Abstract

Cardiomyocytes are one of the most mitochondria-rich cell types in the body, with ∼30–40% of the cell volume being composed of mitochondria. Mitochondria are well established as the primary site of adenosine triphosphate (ATP) generation in a beating cardiomyocyte, generating up to 90% of its ATP. Mitochondria have many functions in the cell, which could contribute to susceptibility to and development of cardiovascular disease (CVD). Mitochondria are key players in cell metabolism, ATP production, reactive oxygen species (ROS) production, and cell death. Mitochondrial calcium (Ca2+) plays a critical role in many of these pathways, and thus the dynamics of mitochondrial Ca2+ are important in regulating mitochondrial processes. Alterations in these varied and in many cases interrelated functions play an important role in CVD. This review will focus on the interrelationship of mitochondrial energetics, Ca2+, and ROS and their roles in CVD. Recent insights into the regulation and dysregulation of these pathways have led to some novel therapeutic approaches.

1. Introduction

Cardiovascular disease (CVD) is the leading cause of death in much of the world. Myocardial ischaemia results in damaged myocardium, and due to the poor proliferative capacity of cardiomyocytes this loss of myocardium results in cardiac hypertrophy, which leads to heart failure. Reperfusion is the main therapy for myocardial ischaemia, but reperfusion per se can initiate reperfusion injury. Mitochondria play an important role in cell death and energetics, which are key regulators of outcomes in ischaemia/reperfusion (I/R) injury and heart failure. In turn, mitochondrial calcium (Ca2+) and reactive oxygen species (ROS) are important regulators of both death and energetics. This review will focus on the roles of mitochondrial Ca2+ and ROS in regulating cell death and mitochondrial energetics and their impact on CVD.

2. Mitochondria and energetics

The role of mitochondria in regulating adenosine triphosphate (ATP) production is well established, and as it has been reviewed in several recent publications,1,2 we will limit our discussion to how mitochondrial energetics are implicated in CVD, namely I/R and heart failure.

2.1 Energetics and I/R injury

During ischaemia the lack of oxygen inhibits cytochrome c oxidase, thereby blocking the flow of electrons in the mitochondrial electron transport chain. As a result of the inhibition of electron flow, Complex I of the electron transport chain is reduced, preventing donation of electrons to Complex I from its substrate NADH, and thus NADH becomes elevated. In a perfused heart model of global ischaemia, the heart continues to beat and consume ATP for a few minutes. Creatine phosphate (CrP) breaks down to regenerate ATP, thereby buffering the fall in ATP. With the cessation of beating, ATP falls gradually until the heart goes into contracture, at which point ATP falls more rapidly3 and the mitochondrial membrane potential also falls at this time.4 Studies by Jennings et al.5 demonstrated that during anoxic conditions, the F1F0-ATP synthase (F1-ATPase) may paradoxically reverse into an energy-consuming enzyme, further favouring H+ extrusion at the expense of ATP hydrolysis. However, for the F1-ATPase to consume cytosolic ATP, the adenine nucleotide transporter (ANT) also is required to function in reverse. Theoretical calculations based on the reversal potential of the F1-ATPase and the ANT raise questions about conditions under which this process occurs.6

Cardiac metabolism during ischaemia depends on the substrates supplied to the heart and the intrinsic metabolic regulation of the heart. To examine the alterations in the intrinsic metabolism of the heart, studies have been done in ex vivo hearts in which the perfusate can be varied. The heart is highly dependent on oxidative metabolism and oxidative phosphorylation by the mitochondria, and yet the heart is omnivorous and can utilize multiple substrates.7 With global ischaemia oxidative metabolism is inhibited and the cell relies on glycolysis to generate ATP (Figure 1). Glucose is metabolized to pyruvate, generating NADH. Conversion of pyruvate to lactate regenerates NAD+, relieving the inhibition of glycolysis by NADH. Acidosis (commonly referred to as lactic acidosis) occurs during ischaemia due to the retention of protons from breakdown of the glycolytically generated ATP.8 This increase in cytosolic protons during ischaemia stimulates H+ exchange with extracellular Na+ on the plasma membrane Na+–H+ exchanger (NHE), thereby raising intracellular Na+ which leads to an increase in cytosolic Ca2+ via the plasma membrane Na+–Ca2+ exchanger (NCX).9 As discussed in Section 4.3, this increase in cell Ca2+ is thought to be a primary trigger of cell death in ischaemia–reperfusion. The increase in NADH and FADH also leads to inhibition of fatty acid oxidation leading to accumulation of acyl carnitines and acyl CoA.10

A simplified illustration of cellular changes during ischaemia. This schematic shows that the absence of oxygen (O2) inactivates Complex IV and the electron transport chain, thus preventing ATP generation through the F1-ATPase. This leads to increased glycolysis and the production of lactate, which acidifies the cytosol. NHE is thus stimulated to exchange H+ ions for Na+, and increased cytosolic Na+ in turn activates NCX, leading to increased cytosolic Ca2+. During ischaemia, mitochondrial Ca2+ is thought to increase. The rise of mitochondrial Ca2+ is canonically attributed to transport of cytosolic Ca2+ into the mitochondrial matrix via MCU, though this has not been directly proven. Overaccumulation of mitochondrial Ca2+ is thought to trigger opening of PTP, which may be inhibited by low pH during ischaemia but could become activated upon reperfusion. PTP opening is thought to lead to cell death.
Figure 1

A simplified illustration of cellular changes during ischaemia. This schematic shows that the absence of oxygen (O2) inactivates Complex IV and the electron transport chain, thus preventing ATP generation through the F1-ATPase. This leads to increased glycolysis and the production of lactate, which acidifies the cytosol. NHE is thus stimulated to exchange H+ ions for Na+, and increased cytosolic Na+ in turn activates NCX, leading to increased cytosolic Ca2+. During ischaemia, mitochondrial Ca2+ is thought to increase. The rise of mitochondrial Ca2+ is canonically attributed to transport of cytosolic Ca2+ into the mitochondrial matrix via MCU, though this has not been directly proven. Overaccumulation of mitochondrial Ca2+ is thought to trigger opening of PTP, which may be inhibited by low pH during ischaemia but could become activated upon reperfusion. PTP opening is thought to lead to cell death.

On reperfusion, oxygen is restored allowing electron transport and oxidative metabolism to resume. If the ischaemic time was short and did not lead to substantial cell death, electron transport resumes generating the mitochondrial membrane potential which in turn generates ATP via Complex V. The relative percentage of ATP generated by oxidation of fatty acids increases due to a decrease in malonyl CoA, an inhibitor of mitochondrial fatty acid transport.11 The increase in fatty acid oxidation leads to inhibition of glucose oxidation, thereby uncoupling glycolysis from glucose oxidation. As discussed above glycolysis generates pyruvate, and if pyruvate is not oxidized by the mitochondria it is converted to lactate. This can lead to an increase in generation of protons and hence via NHE and NCX increased entry of Ca2+ into the cell. Dichloroacetic acid (DCA), which activates pyruvate dehydrogenase (PDH) and thereby activates glucose oxidation has been shown to be protective in I/R injury (Figure 2). The details of altered substrate selection and its role in I/R injury have been discussed in detail elsewhere.12,13

Mitochondrial pathways and potential mitochondrial therapeutic targets in ischaemia/reperfusion (I/R). This schematic illustrates in a simplified manner the interconnections of mitochondrial metabolism, Ca2+, and ROS, and potential therapeutic targets in I/R. The activation of the permeability transition pore (PTP) by Ca2+ and ROS is shown by arrows. Two potential PTP candidates, ANT and the F1-ATPase, are shown. Interventions are shown with gray arrows. Stimulation of PDH with DCA is protective presumably due to enhancing glucose oxidation. Inhibitors of PTP, such as Cyclosporin A (CsA), are also protective. Ca2+ and ROS are activators of PTP and it is proposed that reducing mitochondrial Ca2+, either by blocking mitochondrial Ca2+ uptake through MCU (by Ru360 or recently characterized small molecules) or by enhancing efflux through NCLX (for which there is currently no drug), might be beneficial. Reducing ROS, either by inhibiting SDH (Complex II) and reverse electron transfer (RET), by inhibiting electron leak at Sites I and III, or by mitochondrial targeted antioxidants have all shown benefit. SS-31 which binds to cardiolipin (CL) has also been shown to be beneficial, possibly by modulating the binding of cardiolipin to ANT and potentially inhibiting PTP. Other effects of SS-31, for instance against ROS via interactions of cardiolipin with cytochrome c and the electron transport chain, are not shown for clarity, but likely contribute to SS-31’s efficacy.
Figure 2

Mitochondrial pathways and potential mitochondrial therapeutic targets in ischaemia/reperfusion (I/R). This schematic illustrates in a simplified manner the interconnections of mitochondrial metabolism, Ca2+, and ROS, and potential therapeutic targets in I/R. The activation of the permeability transition pore (PTP) by Ca2+ and ROS is shown by arrows. Two potential PTP candidates, ANT and the F1-ATPase, are shown. Interventions are shown with gray arrows. Stimulation of PDH with DCA is protective presumably due to enhancing glucose oxidation. Inhibitors of PTP, such as Cyclosporin A (CsA), are also protective. Ca2+ and ROS are activators of PTP and it is proposed that reducing mitochondrial Ca2+, either by blocking mitochondrial Ca2+ uptake through MCU (by Ru360 or recently characterized small molecules) or by enhancing efflux through NCLX (for which there is currently no drug), might be beneficial. Reducing ROS, either by inhibiting SDH (Complex II) and reverse electron transfer (RET), by inhibiting electron leak at Sites I and III, or by mitochondrial targeted antioxidants have all shown benefit. SS-31 which binds to cardiolipin (CL) has also been shown to be beneficial, possibly by modulating the binding of cardiolipin to ANT and potentially inhibiting PTP. Other effects of SS-31, for instance against ROS via interactions of cardiolipin with cytochrome c and the electron transport chain, are not shown for clarity, but likely contribute to SS-31’s efficacy.

2.2 Energetics and heart failure

The failing heart has been described as an energy starved heart, and studies have shown that failing human hearts have a decrease in CrP/ATP.14 Depending on the type and severity of heart failure, an increase, decrease, or no change in fatty acid oxidation has been reported.2 In severe heart failure associated with ischaemia or hypertension there is a transition from beta-oxidation to increased glycolysis, accompanied by a return to a more fetal metabolic profile. The increase in glycolysis during heart failure occurs often without an increase in glucose oxidation. Inhibitors of fatty acid oxidation, such as trimetazidine (a ketothiolase inhibitor) as well as etomoxir and perhexiline (CPT1 inhibitors) have been generally shown to be protective in heart failure models, which are proposed to be due, at least in part, to an increase in glucose oxidation (Figure 3). Similarly, inhibitors of malonyl CoA decarboxylase, an enzyme which catabolizes malonyl CoA, leads to an increase in malonyl CoA and thereby decreases fatty acid oxidation because malonyl CoA inhibits fatty acid uptake. Activation of PDH and hence glucose oxidation by DCA has also been shown to be protective in heart failure models.

Mitochondrial pathways and potential mitochondrial therapeutic targets in heart failure. This schematic illustrates in a simplified manner the interconnections of mitochondrial metabolism, Ca2+, and ROS, and potential therapeutic targets in heart failure. As in Figure 2, interventions are shown with gray arrows. Many of the targets are the same as in Figure 2, including PDH, PTP, MCU, and ROS. Activation of PTP is proposed to play a role in heart failure, thus inhibitors of PTP and ROS have also been shown to be beneficial. SS-31 has also been shown to improve heart failure outcomes. In some heart failure models in which mitochondrial Ca2+ is insufficient, inhibiting NCLX with CGP37157 has had efficacy. In heart failure, inhibitors of fatty acid oxidation including CPT inhibitors are protective, and increased ketone metabolism to presumably stimulate glucose oxidation is also beneficial.
Figure 3

Mitochondrial pathways and potential mitochondrial therapeutic targets in heart failure. This schematic illustrates in a simplified manner the interconnections of mitochondrial metabolism, Ca2+, and ROS, and potential therapeutic targets in heart failure. As in Figure 2, interventions are shown with gray arrows. Many of the targets are the same as in Figure 2, including PDH, PTP, MCU, and ROS. Activation of PTP is proposed to play a role in heart failure, thus inhibitors of PTP and ROS have also been shown to be beneficial. SS-31 has also been shown to improve heart failure outcomes. In some heart failure models in which mitochondrial Ca2+ is insufficient, inhibiting NCLX with CGP37157 has had efficacy. In heart failure, inhibitors of fatty acid oxidation including CPT inhibitors are protective, and increased ketone metabolism to presumably stimulate glucose oxidation is also beneficial.

An increase in ketone metabolism has also been shown in heart failure,15,16 and studies were performed to evaluate whether the increase in ketone utilization is adaptive or maladaptive. Recent studies suggest that enhanced ketone oxidation increases energy production but does not improve cardiac efficiency.17 An increase in ketone metabolism can be accomplished by increasing circulating ketones. Interestingly, sodium-glucose transport protein 2 (SGLT2) inhibitors, which were originally given as glucose-lowering drugs have been shown to have additional cardioprotective benefit in heart failure patients in addition to their ability to lower serum glucose.18 Although the exact mechanism by which SGLT2 inhibitors confer this protection is debated, they have been shown to increase circulating ketone levels, which could be involved in the protection. Insulin19 and GLP-120,21 have also been shown to be beneficial. Finally as discussed elsewhere22–24 supplementation with nicotinamide or other NAD precursors has been shown to be protective in heart failure.

3. Mitochondrial Ca2+

3.1 Mitochondrial Ca2+ and energetics

Mitochondrial Ca2+, which is altered in many CVDs, has been shown to regulate ATP production.25 Denton and McCormack26 demonstrated that an increase in cytosolic Ca2+, as occurs with an increase in energy demand, leads to an increase in mitochondrial Ca2+ levels. Ca2+ enters mitochondria through the mitochondrial Ca2+ uniporter (MCU) complex27 and in the matrix activates three mitochondrial NAD(P)H-generating dehydrogenases26: PDH, isocitrate dehydrogenase (ICDH), and α-ketoglutarate dehydrogenase. Ca2+ activation of PDH occurs primarily via Ca2+ activation of PDH phosphatase, which results in dephosphorylation and activation of PDH. Ca2+ activation of these dehydrogenases stimulates increased NADH production, which provides electrons to the electron transport chain at Complex I to enhance ATP production. Brandes and Bers28 showed in cardiac trabeculae that with an increase in pacing, there was a rise in mitochondrial Ca2+ and transient elevation of NADH. Liu and O’Rourke29 studied adult guinea pig cardiomyocytes and found that the relationship between an increase in mitochondrial Ca2+ and a change in NADH with increased pacing was dependent on cellular Na+ concentration. If the experiment was done in the presence of 15 mM Na+ (compared with 5 mM Na+), upon pacing there was less of an increase in mitochondrial Ca2+ and an oxidation in NADH, suggesting that Na+-dependent mitochondrial Ca2+ efflux plays a role in regulating mitochondria Ca2+ during increased work. In the presence of Ru360, an inhibitor of MCU, the pacing-induced increase in mitochondrial Ca2+ was reduced and net NADH oxidation was observed.

In addition to Ca2+ activation of the mitochondrial dehydrogenases, Ca2+ has also been suggested to activate mitochondrial electron transport Complexes III and V.25 However, Wescott et al.30 recently reported that an increase in matrix Ca2+ only activates ATP production from NADH-linked substrates via Ca2+-activated dehydrogenases, thereby questioning Ca2+ activation of Complex III or V. Additional studies will be needed to resolve this discrepancy.

3.2 Mitochondrial Ca2+ and cell death pathways

Cardiac cell death can be initiated by multiple pathways and the relative importance of these pathways depends upon a number of context-dependent factors. Autophagy, mitophagy, and ferroptosis can initiate cell death and are reviewed elsewhere.31–34 This review will focus on Ca2+ activation of necroptosis and opening of the mitochondrial permeability transition pore (PTP). Under pathological conditions of Ca2+ overload, Ca2+ can enter mitochondria presumably via the MCU leading to an increase in mitochondrial Ca2+ and opening of the PTP.35 The PTP is a large conductance channel in the mitochondrial inner membrane. Studies in isolated mitochondria showed that cyclosporin A (CsA) can inhibit Ca2+-activated PTP opening and the resultant mitochondrial swelling. Cyclophilin D (CypD), a mitochondrial matrix protein, was shown to be the target of CsA. An increase in matrix Ca2+ and ROS, which both occur with ischaemia and reperfusion, triggers PTP opening. Sustained opening of PTP leads to loss of mitochondrial cofactors and proteins up to ∼1.5 kDa. Thus, PTP opening leads to loss of mitochondrial membrane potential and loss of mitochondrial cofactors such as NAD/NADH. Low pH inhibits PTP opening.36,37 However, this effect is complex, as low pH inhibits PTP opening only in de-energized mitochondria.37

Although CypD has been shown to be a regulator of the PTP, it is not the pore-forming unit. Unfortunately, the identity of the PTP is still debated, limiting progress on devising treatments to block PTP opening during I/R. There are a number of recent reviews on the PTP38–41 so that topic will not be discussed in detail here. Briefly, recent data have suggested that the F1F0-ATP synthase (F1-ATPase) can, under certain circumstances, form the PTP. Bernardi’s group has proposed that dimers of the F1-ATPase can form the pore-forming unit of the PTP.42 Jonas’ group has suggested that delipidation of the c-ring of the F1-ATPase can form the PTP.43 Although there are strong data in support for both hypotheses, there are also data in opposition. Data from Walker’s group have shown a CsA-inhibitable PTP-like swelling is still present in cells lacking the c-ring and other components of the F1-ATPase that are proposed to be involved in PTP formation.44,45 One explanation for the discrepancy is that there are two or more CsA-inhibitable pores. Data from over 30 years ago proposed a role for ANT as a key component of PTP.46,47 CypD was shown to bind to ANT and adenine nucleotides were known inhibitors of PTP. Bongkrekic acid (BKA) and carboxyatractyloside (CA), two inhibitors of ANT that lock ANT in different conformations, were shown to regulate PTP.47 BKA locks ANT in the matrix facing conformation and inhibits PTP, whereas CA binds to the cytosolic facing conformation and activates PTP. The hypothesis that ANT was the PTP fell out of favour in 2005 when Wallace’s group reported that a CsA-inhibitable PTP, albeit requiring higher Ca2+ for activation, was present when ANT1 and 2 were deleted.48 Recent studies have revisited a role for ANT as a pore component of PTP.49 Karch et al.49 showed that mitochondria with deletion of ANT1, 2, and 4 were desensitized to Ca2+-induced PTP opening, and PTP opening was completely inhibited by the further addition of CsA. They proposed that there are two distinct PTPs: the ANT and a second pore. Both components may be activated by CypD, although the non-ANT component has a strict requirement for CypD.

Although a role for Ca2+ in PTP opening has been clearly demonstrated, the mechanism by which Ca2+ activates PTP is unclear. Much of the difficulty with deciphering how Ca2+ activates PTP is related to the lack of clarity regarding the identity of PTP. Ca2+ binding to T163 of the beta-subunit of the F1-ATPase alters its conformation to promote pore opening at the dimer/tetramer interface of the Fo domain.50 Mutation of T163 alters Ca2+- and Mg2+-dependent ATP hydrolysis. It was further demonstrated in HeLa cells that when T163 is mutated, the Ca2+ sensitivity of the PTP is attenuated and almost twice as much Ca2+ is required to activate PTP.50

Moreover, Ca2+ can bind to cardiolipin, which is known to be in close proximity with several major transport proteins including ANT. It has been proposed that Ca2+ binding to cardiolipin might regulate the conformation of ANT and thus promote PTP.51 Alternatively, glutamate 152/3 and aspartate 167/8 of ANT have been proposed to play a role in regulating Ca2+ binding to ANT to promote PTP47; however, this has not been confirmed experimentally.

CypD, a key activator of PTP, has been shown to possess an acylation site at C202. Acylation is the attachment of a fatty acid to a cysteine via a thioester bond. Interestingly, high Ca2+ has been shown to lead to de-acylation of CypD at C202.52 It is proposed that de-acylation of CypD might be important in its targeting to PTP; perhaps acylation targets it away from PTP or alternatively a free C202 might be important to activate PTP. Clearly, conclusive identification and better mechanistic understanding of PTP will be important in elucidating the mechanism by which Ca2+ and other regulators of the PTP mediate their effects.

Mitochondrial Ca2+ may influence cell death and survival by mechanisms that go beyond triggering of PTP. One of these mechanisms could be the activation of mitochondrial calpains, which have been shown to play a role in cardiomyocyte injury and cell death.53,54 In addition, mitochondrial Ca2+ uptake during I/R could buffer cytosolic Ca2+ overload, a trigger of reperfusion-induced hypercontraction and death. Indeed, in certain conditions such as ageing in which mitochondrial Ca2+ uptake is reduced or impaired cytosolic Ca2+ overload is aggravated, and this effect is associated with an increased cardiomyocyte hypercontracture and sarcolemmal rupture.55

3.3 Mitochondrial Ca2+ at baseline and during ischaemia and reperfusion

3.3.1 Baseline pre-ischaemic Ca2+

There is some variability in measurements of the baseline level of mitochondrial Ca2+, but in general it is reported to be 100–200 nM or lower.56–58 Andrienko et al.56 reported that for cytosolic free Ca2+ levels at or below 475 nM, mitochondrial Ca2+ is lower than cytosolic Ca2+. As discussed elsewhere, there has been some disagreement about whether mitochondrial Ca2+ can respond on a beat-to-beat basis or whether mitochondrial Ca2+ acts to integrate increases in the cytosolic Ca2+ transient.59,60

3.3.2 Ca2+ during ischaemia

Studies done over two decades ago showed that an increase in cellular Ca2+ was an important trigger of necrotic cell death.9 As mentioned above, it was shown that during myocardial ischaemia there is an increase in acidosis that promotes plasma membrane NHE activity, leading to an increase in cytosolic Na+, which in turn leads to an increase in cytosolic Ca2+ via NCX9 (Figure 1). The importance of these ionic changes in triggering cell death was demonstrated by showing that inhibitors of NHE attenuated the rise in Na+ and Ca2+ during ischaemia and were cardioprotective.9 However, these NHE inhibitors were not protective in a large clinical trial.61 Reasons for the lack of protection are discussed elsewhere62 and include an increase in stroke. However, the lack of protection is consistent with the preclinical data, which showed that the rise in Na+ and Ca2+ occur during ischaemia, before patients can be treated.9 Although a number of drugs have been reported in animal studies to be beneficial when added on reperfusion,63,64 few if any have shown benefit in clinical trials. Possible reasons for this have been discussed elsewhere.65,66 The field has shifted focus to try to understand the downstream pathways by which cytosolic Ca2+ initiates cell death. One current hypothesis is that during ischaemia and reperfusion Ca2+ enters the mitochondria via the MCU to activate the PTP. The PTP is reported to be inhibited by acidic conditions that are present during ischaemia and is therefore not activated until reperfusion when the pH is restored.67 The inhibition of PTP during ischaemia provides a window of opportunity to get PTP inhibitors to cells at the start of reperfusion to inhibit PTP opening. Addition of CsA or deletion of CypD has been shown to reduce cell death following I/R in isolated heart68 and animal studies.69,70 An initial small clinical trial demonstrated CsA protection in acute myocardial infarction (MI)71; however, a recent clinical trial did not find CsA to protect in patients with MI.72 A possible reason could be that CsA does not inhibit the PTP itself; rather, it inhibits CypD, an activator of the PTP. It has been shown that with higher Ca2+ or ROS levels, PTP opening can occur in the presence of CsA.73–76 Thus, future development of direct inhibitors of the PTP itself could potentially be beneficial.

An alternative to inhibiting PTP is to block the upstream increase in mitochondrial Ca2+. As MCU is the only documented mitochondrial Ca2+ import channel,77,78 it has been suggested that inhibiting MCU in I/R might be a strategy to reduce PTP opening.79 It should be noted that if mitochondrial Ca2+ increases during ischaemia this would limit the protective effect of MCU inhibitors added at the start of reperfusion. To test the role of MCU in regulating mitochondrial Ca2+ in CVD, several lines of mice with MCU deletion were developed. In the first model, MCU was deleted globally using a gene trap method in the germline80 (Mcu–/– mice). In another model MCU was conditionally deleted in adult cardiac cells using Mcu floxed mice crossed with the tamoxifen-inducible α-MHC-driven Mer-Cre-Mer81,82 (Mcufl/fl–MCM mice). Cardiac mitochondria from both lines were studied and shown not to take up Ca2+ or undergo PTP. An α-MHC-driven dominant negative MCU (DN-Mcu) was also developed,83,84 and similar to the Mcu–/– and Mcufl/fl–MCM mitochondria, mitochondria from this mouse did not take up Ca2+ and did not undergo PTP.

As mitochondria from the Mcu–/–, Mcufl/fl–MCM, and DN-Mcu mice do not take up Ca2+ and do not undergo PTP, it was expected that they would be protected from I/R. Surprisingly, there was no protection following I/R in the germline Mcu–/– mice,80 and CsA provided no protection in the Mcu–/– mice. Furthermore, there was no protection in the mice with cardiac specific expression of DN-Mcu.83 In contrast, when MCU was conditionally deleted in adult cardiac cells, the hearts were protected from I/R injury.81,82 The reason for this discrepancy is not clear, but it has been proposed that adaptive changes are likely to occur when MCU activity is lost (by deletion or a DN) at or before birth.85 The precise nature of these adaptations is under study, but it appears that in the germline Mcu–/– mitochondria less Ca2+ is required for PTP activation.85 It is also unclear whether the adaptations are related to development or whether they would occur in adult mice with additional time after acute MCU deletion. As mentioned above, CsA provides no protection in the germline Mcu–/– mice80; perhaps the adaptations, which occur in the Mcu–/– mice might provide some insight into why CsA did not provide protection in a recent clinical trial.72

It should also be noted that the studies showing protection in the adult Mcufl/fl–MCM hearts did not directly confirm that the protection was due to blocking an increase in mitochondrial Ca2+ during I/R.81,82 A recent abstract reported that simulated ‘ischaemia’ in neonatal mouse cardiomyocytes isolated from the conditional Mcufl/fl mouse and treated with Cre-expressing adenovirus to delete MCU led to similar mitochondrial Ca2+ increases as in control cardiomyocytes.86 Clearly, additional studies are needed to fully address the role of mitochondrial Ca2+ in I/R. The development of new methodology to measure mitochondrial Ca2+ in beating perfused hearts should be helpful in addressing this question.87

Inhibition of MCU to block the rise in mitochondrial Ca2+ and hence activation of PTP is proposed to be beneficial in I/R. Therefore, drugs targeting MCU might be beneficial in CVD, though existing inhibitors of MCU such as Ru360 are limited in membrane permeability. Thus, recent screening has identified new small molecule inhibitors of MCU activity.79 However, one such molecule, MCU-i11, was shown to be ineffective against cell death in a cardiomyocyte-based in vitro hypoxia/reoxygenation model79 and was suggested to potentially be of use in chronic mild Ca2+ overload conditions, though this remains to be tested.

In addition to the MCU-mediated Ca2+ uptake pathway, there are Ca2+ efflux pathways.88,89 In steady state, the Ca2+ that enters mitochondria through the MCU must exit via a Ca2+ efflux pathway. Data from the 1970s suggested that there are Na+-dependent and Na+-independent Ca2+ efflux mechanisms. The protein responsible for mitochondrial Na+-dependent Ca2+ efflux (Na+–Ca2+–Li+ exchanger; NCLX) was identified in 2010.89 NCLX is thought to be electrogenic; therefore, in the presence of a typical large negative mitochondrial Δψ, it will only function to extrude Ca2+. A mouse with conditional deletion in NCLX in adult heart was developed and used to show that acute loss of NCLX was associated with high mortality as most mice died within days after NCLX deletion.90 These data suggest that NCLX is a major pathway for Ca2+ efflux in the heart. Mice with cardiac specific overexpression of NCLX were generated and it was shown that hearts from these mice are protected from I/R injury.90 These data are consistent with the concept that increased NCLX-dependent Ca2+ efflux during I/R reduces mitochondrial Ca2+ and can mediate cardioprotection.

3.3.3 Mitochondrial Ca2+ during reperfusion

What happens to mitochondrial Ca2+ on reperfusion depends on a number of factors with the length of ischaemia being an important factor. In perfused rat heart after a short (20–25 min) period of ischaemia, cytosolic Ca2+ returns close to baseline Ca2+ by ∼30 min of reperfusion.9 It should also be noted that the length of ischaemia that leads to more irreversible changes varies depending on the species. Studies in isolated cardiomyocytes report oscillations in cytosolic Ca2+ on reperfusion, which appear to be due to oscillations in sarcoplasmic reticulum (SR) Ca2+ release and re-uptake. As reviewed elsewhere,91,92 a number of mechanisms were identified by which cytosolic Ca2+ can modulate injury on reperfusion. There are few if any reports of how mitochondrial Ca2+ responds during reperfusion. It might be expected to follow the changes in cytosolic Ca2+ with perhaps some lag. Future studies will be needed to address the changes that occur in mitochondrial Ca2+ on reperfusion. As mitochondrial Ca2+ can regulate metabolism and ATP production, prolonged alterations in mitochondrial Ca2+ could have important metabolic and energetic effects.

3.4 Mitochondrial Ca2+ and heart failure

A great deal of controversy exists surrounding to what degree mitochondrial Ca2+ uptake increases or decreases in the progression of heart failure. Some of the discrepancies in the results can be attributed to different animal species (for instance, mouse vs. guinea pig) and different types of induced heart failure. Studies have used various models, including transverse aortic constriction (TAC), post-MI heart failure, or isoproterenol-induced heart failure.

Among the studies pointing to excessive mitochondrial Ca2+ in heart failure, one study showed that elevated mitochondrial Ca2+ levels caused by Ca2+ leak from the SR contributes to heart failure.93 The mouse model of post-MI heart failure examined in this study was induced by permanent occlusion of the proximal left anterior descending (LAD) coronary artery. Direct measurement of mitochondrial Ca2+ levels revealed a more than two-fold increase in post-MI heart failure samples relative to sham. Concurrently, SR Ca2+ leak and production of ROS increased in post-MI isolated cardiomyocytes. Mice harbouring a constitutively leaky mutation of the Type 2 ryanodine receptor (RyR2) channels (termed RyR2-S2808D mice) were used to demonstrate that the SR Ca2+ leak causally increased mitochondrial Ca2+, resulting in dysmorphic and malfunctioning mitochondria as well as loss of membrane potential. RyR2-S2808D mice exhibited further elevation of mitochondrial Ca2+ levels in sham and post-MI conditions, and these mice also showed exacerbated progressive decline in ejection fraction measured in the weeks post-MI. Although this study examined only post-MI heart failure, it overall supports the view that increased mitochondrial Ca2+, in this case due to SR Ca2+ leak, contributes to the progression of heart failure. A similar mechanism for increased SR Ca2+ leak leading to mitochondrial Ca2+ overload has been shown to occur with ageing in both mice and humans.94

In a study mentioned above, elevated mitochondrial Ca2+ induced by blocking export via NCLX acutely in adulthood led to severe heart defects and a high rate (87%) of spontaneous death.90 Tamoxifen-induced, heart-specific deletion of Slc8b1, the gene encoding NCLX, in adult mice led to hallmarks of heart failure including left ventricular (LV) dilation and decreased fractional shortening, hypertrophy, and increased fibrosis.90 These effects were attributed to opening of the PTP, a known consequence of mitochondrial Ca2+ overload, and could be rescued by deletion of CypD, an activator of PTP opening. As discussed above, transgenic overexpression of NCLX in the heart increased Ca2+ efflux from the mitochondria and protected against I/R injury.90 In addition, overexpression of NCLX also reduced pathological heart remodelling after permanent occlusion of the LAD coronary artery, the same post-MI heart failure model used in the above-mentioned study.93 Interestingly, increased mRNA expression of SLC8B1 was observed in LV tissue of patients with ischaemic heart failure, which the authors suggest represents a compensatory mechanism during heart failure to limit pathological mitochondrial Ca2+ overload. Taken together, these studies suggest that increased mitochondrial Ca2+ is a driver of heart pathology and that decreasing mitochondrial Ca2+ would be a potential therapeutic avenue.

Results using TAC rather than post-MI heart failure in mice have been unclear on the utility of deleting MCU. In both a global knockout mouse model95 and cardiac-specific tamoxifen-inducible mouse model,81 MCU deletion did not alter the course of pathology after TAC. However, it is not completely clear whether MCU deletion can prevent the chronic accumulation of mitochondrial Ca2+; without MCU, the acute Ca2+ response to adrenergic stimulation seems to be blunted,87 but in pathological or chronic conditions, more investigation is needed. In fact with 60 min of adrenergic stimulation an increase in mitochondrial Ca2+ was noted in the cardiac-specific tamoxifen-inducible Mcufl/fl–MCM mouse model.81 There is evidence, however, that germline deletion of MICU3, a regulator of the uniporter that enhances mitochondrial Ca2+ uptake, attenuates isoproterenol-induced hypertrophy.96 Post-isoproterenol, the Micu3–/– mice concurrently displayed increased phospho-PDH to total PDH ratio relative to wild-type controls, indicative of lower mitochondrial Ca2+. Hence, limiting mitochondrial Ca2+ uptake remains a potential avenue to ameliorate heart failure.

Another substantial body of evidence supports a role for decreased mitochondrial Ca2+ in worsening heart failure. A number of studies have proposed that heart failure-associated changes in intracellular Ca2+ and Na+ handling lead to decreased mitochondrial Ca2+, impairing mitochondrial energy production and depleting mitochondrial antioxidative capacity. Mimicking heart failure conditions including reduced systolic SR Ca2+ release by patch clamp in guinea pig myocytes, one study showed that MCU activity decreased and failed to stimulate Ca2+-sensitive Krebs cycle enzymes.97 Consistent with these results, blocking NCLX with the inhibitor CGP-37157, thus presumably increasing mitochondrial Ca2+, prevented imbalance in energetics (NADH oxidation).29 Indeed, chronic CGP-37157 in a guinea pig model of heart failure and sudden cardiac death (SCD) using aortic constriction and adrenergic receptor stimulation was beneficial and attenuated hypertrophic remodelling and dysfunction.98 Further in support of the view that heart failure is linked to depletion of mitochondrial Ca2+, increased intracellular Na+ concentration in heart failure was shown to drive increased NCLX activity, accelerating Ca2+ export from the mitochondria.99 The consequences of reduced mitochondrial Ca2+ are thought to be not only bioenergetic impairment but also increased ROS. Because regeneration of NADPH, an important component of mitochondrial antioxidant capacity, relies on the Krebs cycle, lower mitochondrial Ca2+ depletes NADPH and leads to increased oxidative stress. Again in guinea pig myocytes, NCLX inhibition via CGP-37157 also prevented increased ROS formation.100 More recently, a study showed that consistent with these results, overexpression of MCU increased mitochondrial Ca2+, decreased ROS, and improved contractility in a guinea pig model of hypertrophy via aortic banding and isoproterenol.101

It should also be noted that cardiac myocytes have several mitochondrial populations (subsarcolemmal, interfibrillar, perinuclear), with distinct energetic traits and Ca2+ tolerance properties. These traits may be relevant in pathological conditions, such as heart failure and I/R injury. On the other hand, subsarcolemmal mitochondria differentially contain connexin 43 (Cx43)102 that has been shown to have a pathophysiological role in I/R injury.103

Summed together, the evidence suggests there may be an optimal physiological range for mitochondrial Ca2+ and that straying either too low or too high is detrimental.104 Further studies are needed to determine under what circumstances mitochondrial Ca2+ increases or decreases excessively, and whether a change in either direction is a cause or effect of heart failure. Our current understanding does not exclude the possibilities that elevation or decline in mitochondrial Ca2+ could be associated with different types of heart failure or that during different stages of heart failure mitochondrial Ca2+ shifts from one extreme to the other.

4. Mitochondrial ROS

4.1 Mechanisms involved in regulating ROS

ROS can lead to oxidative stress and are inextricably linked to mitochondrial function, as oxidative phosphorylation inherently generates ROS as by-products. Oxidative stress is defined as an imbalance of ROS production and scavenging. Electron leaks from at least 11 distinct sites in the electron transport chain can generate ROS in the form of superoxide anions (O2) and hydrogen peroxide (H2O2).105 The capacities of these sites to produce O2 and H2O2 vary substantially,105 and the capacity of a given site can vary among tissues106 and even between cardiac subsarcolemmal and interfibrillar mitochondria.107 Canonically, Complexes I and III are recognized as the primary sites of mitochondrial production of ROS,108,109 but under different substrate and exercise conditions other sites also contribute.110 In addition to sites within the electron transport chain, several other mitochondrial proteins associated with the inner and outer mitochondrial membranes can contribute to ROS production,111 including p66Shc, monoamine oxidase isoforms A and B (MAO-A and MAO-B), and NADPH Oxidase 4 (NOX4).

The impact of mitochondrial ROS on cellular and heart function is complex. Though evidence exists that ROS can also serve as molecular signals112 as will be discussed later, ROS cause damage to macromolecules including proteins and DNA. Thus, it is generally accepted that oxidative stress plays an important pathological role in CVDs.113 For instance, ROS overload is thought to contribute to opening of the PTP, which as mentioned above is damaging to mitochondria and cells. In a phenomenon known as ROS-induced ROS release, opening of the PTP upon ‘triggering’ ROS leads to a further burst of ROS, thus increasing oxidative stress and leading to more damage.114,115

The chemical reactivities of O2 and H2O2 are different, and thus they target distinct types of molecules.116 Superoxide is highly reactive and oxidizes positively charged Fe–S centres in proteins including the TCA cycle enzyme aconitase.117 Hydrogen peroxide is reactive with thiolates, such as cysteine residues on PDH Kinase 2, and being membrane-permeant can also diffuse to the cytosol and impact kinase and transcription factor signalling.112 To mitigate ROS-mediated oxidative stress, cells employ antioxidant defence systems that consume O2 and H2O2. O2 can be converted into H2O2 by superoxide dismutases (SOD family), of which one member is mitochondrial (SOD2, also known as Mn-SOD), or by spontaneous dismutation. Conversion of H2O2 to water in the mitochondrial matrix can be mediated by matrix thiol systems, including members of the peroxiredoxin (Prx) family, such as mitochondrial Prx3, and the glutathione peroxidase (Gpx) family.118 Prx and Gpx rely on the cofactors thioredoxin and glutathione, respectively, which require NADPH for regeneration. The peroxisomal protein catalase can also consume H2O2, and a limited number of studies show evidence that it can localize to mitochondria in cardiomyocytes.119,120 Despite some controversy in this regard, mitochondria-targeted catalase is nonetheless a useful experimental tool to link matrix H2O2 to a phenotype.116 These ROS scavenging systems each play important roles in maintaining cell function and experimental loss or gain of function can be instructive in differentiating between the contributions of O2 and H2O2 to various pathologies.

It is difficult to define a general set of conditions that necessarily lead to increased ROS generation. Rates of electron leak to O2 or H2O2 do not depend on respiration rate per se but on whether the particular site becomes more oxidized or reduced when electron flow into or out of that site changes.105 Hence, increased respiration rate can lead to either increased or decreased ROS production: if the increased respiration rate is driven by increased substrate supply, further reducing a site, ROS production at that site will increase, but if driven by increased ATP demand or proton leak, oxidizing the site, ROS production will decrease. Indeed, ATP synthase was hyperacetylated in heart failure patients, which was associated with decreased activity.121 In principle, decreased ATP synthesis would pose a roadblock to electron flow and tend to increase ROS production in failing hearts. Even so, identifying the exact sites and mechanisms contributing to increased ROS production in CVDs is complicated. Therefore, many studies rely on methods to scavenge ROS and assess if they mitigate pathological outcomes to help identify whether O2 or H2O2 are responsible.

4.2 ROS and I/R injury

Many studies demonstrate an important role for ROS in I/R injury. Hearts from SOD2+/– mice showed impaired functional recovery in a Langendorff perfusion model of I/R,122 and correspondingly, SOD2 overexpression in transgenic mice was protective both in ex vivo and in vivo I/R models.123 These data indicate that mitochondrial O2 contributes to tissue damage. Interestingly, in simulated I/R in embryonic chick cardiomyocytes, death was attenuated by overexpression of SOD2, but not by catalase, mitochondrial expressed catalase (mCAT), or primarily cytosolic SOD1.124 While more studies are needed, ideally in vivo in a mammalian model, this suggests that O2 might play a more significant role than H2O2 in I/R injury.

I/R has also been studied using other means of targeting the generation of ROS, including the use of small molecules that suppress electron leak to oxygen without inhibiting electron transport in the respiratory chain. Named S1QELs (suppressors of site IQ electron leak) and S3QELs (suppressors of site IIIQo electron leak), these molecules selectively decrease formation of O2 and possibly H2O2 at specific sites.110 Discovered via high-throughput screening, S1QEL1.1 improved cardiac functional recovery and decreased infarct size in a Langendorff-perfused mouse heart model of I/R injury.125 Additionally, the drug OP2113 was shown to act as an S1QEL, and in ex vivo Langendorff-perfused rat hearts OP2113 also improved recovery of contractile performance and reduced infarct size.126 OP2113 was also effective in a rat in vivo post-MI I/R model. When given preventively prior to coronary artery occlusion, OP2113 significantly reduced infarct size.127

The importance of O2 and Complex I in I/R injury is consistent with a recently elucidated metabolic pathway linking I/R to ROS generation. Comparative metabolomics identified accumulation of succinate during ischaemia in multiple metabolically diverse tissues.128 Simulations predicted that upon reperfusion, succinate would rapidly be oxidized by succinate dehydrogenase (SDH; Complex II), driving reverse electron transport through Complex I and generating O2. In line with this hypothesis, infusion of dimethyl malonate to inhibit SDH prevented a burst of mitochondrial ROS at the onset of reperfusion and reduced infarct size after in vivo MI-induced I/R injury in mice128 and in pigs.129 While this model of the relationship between I/R and ROS is not without controversy,130 it fits with the above observations of a critical role in I/R for O2 generated by Complex I.

Additionally, though its mechanism of action against ROS is less direct, the mitochondria-targeted compound SS-31 (Elamipretide, Bendavia) also has been shown to have efficacy in I/R. SS-31 binds to cardiolipin, a phospholipid enriched in the mitochondrial inner membrane, by electrostatic and hydrophobic interaction. Cardiolipin interacts with ANT and the electron transport complexes and has been reported to play a role in cristae structure. Interaction of cardiolipin with cytochrome c has been reported to regulate the cytochrome c switch between an electron carrier and a peroxidase. Cardiolipin oxidation occurs in many CVDs, and Barth syndrome is characterized by loss of the tafazzin gene, which is needed to produce cardiolipin. Loss or oxidation of cardiolipin can alter bioenergetics, ROS, and redox and mitochondrial transporters. By binding cardiolipin, SS-31 has been shown to have beneficial effects, including against I/R injury. SS-31 was protective and reduced infarct size in ex vivo and in vivo I/R models in a range of animals, including mice, rats, guinea pigs, rabbits, and sheep.131–134 Notably, however, when SS-31 was administered to rabbits even 10 min after the onset of reperfusion in vivo, protection was lost.134 Furthermore, even though SS-31 was administered more than 15 min prior to reperfusion, the EMBRACE STEMI trial in 118 patients with MI revealed that SS-31 did not reduce estimated infarct area or produce improvements in clinical outcomes.135

4.3 ROS and heart failure

Multiple studies deleting or decreasing SOD2 have linked O2 to cardiac disease outcomes. SOD2 homozygous deficiency in multiple mouse models led to postnatal death at differing ages and degrees of severity of dilated cardiomyopathy depending on genetic background.136–138 Heart and muscle specific knockout of SOD2 led to hypertrophy, dilatation, and reduced contractility, which were partially rescued by the SOD mimetic MnTBAP.139 In mice heterozygous for SOD2, though myocardial ultrastructural damage was not always observed, cardiomyocytes were shown to be sensitized towards PTP opening.140,141 Strikingly, in humans a genetic defect in SOD2 was associated with severe neonatal cardiomyopathy.142 Conversely, overexpression of SOD2 in mice increased ejection fraction.143 Experiments using the SOD mimetic mitoTEMPO to rescue heart failure phenotypes also provide good evidence that O2 is involved in pathology. In guinea pigs undergoing ascending aortic constriction/beta-adrenergic stimulation to induce nonischaemic heart failure, accompanied by SCD, mitoTEMPO reduced ROS production and prevented and reversed heart failure and SCD.144 Additionally, in mice, pressure overload via TAC induced decreased contractility, which improved with infusion with mitoTEMPO.145 Taken together, all these lines of evidence point to O2 contributing significantly to heart failure.

Mitochondrial targeted antioxidants such as MitoQ have been developed to reduce mitochondrial-generated ROS. In animal studies, MitoQ has shown great promise in reducing both I/R injury146,147 and heart failure.148–150 Although MitoQ showed no improvement over placebo in a patient study for Parkinson’s disease,151 a small human trial for hepatitis C did show a reduction in markers of liver damage in the MitoQ-treated patients.152 It should be mentioned that ROS also has signalling roles in the heart and that prolonged treatment with drugs such as MitoQ could interfere with beneficial ROS signalling. For example, it has been shown that antioxidants can block cardioprotective signalling, as will be discussed below.

Analogous to targeting SOD2 to focus on O2, studies targeting PRDX3, or using mCAT have implicated H2O2 damage in multiple contexts. Transgenic expression of the rat Prx-3 gene in a mouse post-MI heart failure model attenuated MI-induced dilatation of the LV and contractile dysfunction.153 In a pressure overload mouse model using TAC, ROS and protein carbonylation increased and led to decline in contractility, which was partially rescued by expression of mitochondrial catalase.145,154 Taken together, PRDX3 or mCAT rescue of multiple heart failure models strongly demonstrate a causal role for H2O2 in pathologies resembling heart failure with reduced ejection fraction (HFrEF).

The beneficial effects of mitochondrial suppression of ROS are not limited to HFrEF. One study showed that mice fed a high-fat, high-sucrose (HFHS) diet developed cardiac diastolic dysfunction while maintaining systolic function, resembling conditions of heart failure with preserved ejection fraction (HFpEF). Expression of mCAT in transgenic mice ameliorated HFHS diet-induced diastolic dysfunction and prevented an increase in the rate of mitochondrial H2O2 production.155 Hence, while studies are scarcer, combating H2O2 may also prove to be therapeutically beneficial in HFpEF. In further contexts, mCAT has been shown to reduce cardiac disease in aged mice156 and a progeroid mouse model.157 Ageing seems to involve not only H2O2 but O2 as well, as mitoTEMPO also improves systolic and diastolic function in old mice158 and rats.159 Treatment with mitoTEMPO decreased oxidative stress, cell death, and hypertrophy, and led to improved function in diabetic cardiomyopathy.160 Conditions leading to oxidative stress also can lead to glycation of RyR94 that can contribute to the development of heart failure.

The results of SS-31 intervention in the context of heart failure have been more encouraging than the results in I/R. In animal experiments, SS-31 also improved outcomes in multiple heart failure models including TAC161 and angiotensin II treatment162 in mice and in a canine heart failure model.163 Recently, SS-31 appeared to improve cardiac function in a double-blind and placebo-controlled trial of human patients presenting with HFrEF.164 These results provide considerable motivation for moving SS-31 into the clinical setting, though challenges in translating promising results in animal studies to human patients remain.

Collectively these studies, by manipulating various antioxidant proteins, have demonstrated that ROS-mediated damage is a major contributor to the development of heart failure. Alongside these data, a series of studies has explored the molecular underpinnings of oxidative stress observed in heart failure.165 It was observed that in heart failure, rising cytosolic Na+ leads to decreased mitochondrial Ca2+, thus depressing Krebs cycle flux.100 As a result, because reducing NADP+ to NADPH depends on substrates derived from the Krebs cycle, the NADPH pool declines, inhibiting the regeneration of glutathione and thioredoxin, which are required for H2O2 elimination. Overall, the antioxidant capacity of mitochondria decreases, leading to oxidative stress. Furthermore, nicotinamide nucleotide transhydrogenase (NNT), which normally regenerates NADPH from NADH, was reported to work in reverse in conditions of increased afterload and insufficient mitochondrial Ca2+. The reversal of NNT to regenerate NADH for ATP production at the expense of ADP leads to oxidative stress, PTP activation, and cell and tissue damage.166 Overall, targeting the multiple pathways involved in increased ROS generation and decreased ROS scavenging in heart failure could be a promising therapeutic approach.

Despite the abundance of evidence for the negative effects of ROS in CVD, pushing to eliminate ROS entirely can also be harmful. Studies have shown that increasing glutathione pharmacologically or genetically can paradoxically increase oxidation and lead to cell death.167 This phenomenon known as reductive stress, in which excess reducing equivalents induce deleterious effects, has been linked to cardiomyopathy.168 Moreover, ROS could be essential signalling molecules in the heart as they are in other tissues,169 and heightened suppression of ROS could disrupt these signalling pathways. One study found that accumulation of damaged mitochondria, leading to heart failure, in mice lacking cardiac expression of Mitofusin 2 could be rescued by moderate levels of mCAT, but not high levels.170 High mCAT expression was not beneficial because it prevented ROS-mediated activation of autophagic mitochondrial quality control pathways.170 These data support the theory that ROS contribute to mitohormesis, a term referring to mild mitochondrial stresses than can induce a cytoprotective state that is beneficial in the long term.171 Blocking of mitohormetic responses could be a possible explanation for the lackluster results of global antioxidants in treating CVD.111 Indeed, in one study, vitamin E was associated with an increased risk of heart failure in patients with vascular disease or diabetes.172 Overall, although excess ROS are pathological, low levels of ROS can prevent reductive stress and stimulate mitohormesis, thereby providing functional value to the heart.

5. Summary and conclusion

The unique reliance of the heart on mitochondria for energy production puts an interconnected network of mitochondrial functions front and centre in cardiomyocyte health and disease. Mitochondria house multiple metabolic pathways, as well as the electron transport chain. Mitochondrial Ca2+ impinges on the Krebs cycle and the electron transport chain, and it also is a primary activator of the PTP. Electron transport generates ROS, which in addition to causing oxidative stress can also contribute to PTP activation. The regulation of all these mitochondrial pathways are critical for cardiomyocyte homeostasis, and thus it comes as no surprise that their dysregulation is associated with CVD, including I/R injury and heart failure. We have reviewed a growing number of studies that demonstrate the links between various mitochondrial functions and CVD, as well as some approaches that seek to target them for therapeutic benefit. The pace of research in this field promises innovative mitochondria-targeted interventions in CVD in the near future.

Funding

J.C. Liu was supported by K22HL137901 from the National Institutes of Health. EM was supported by the intramural program of NHLBI-NIH.

References

1

Tian
R
,
Colucci
WS
,
Arany
Z
,
Bachschmid
MM
,
Ballinger
SW
,
Boudina
S
,
Bruce
JE
,
Busija
DW
,
Dikalov
S
,
Dorn
GW
,
Galis
ZS
,
Gottlieb
RA
,
Kelly
DP
,
Kitsis
RN
,
Kohr
MJ
,
Levy
D
,
Lewandowski
ED
,
McClung
JM
,
Mochly-Rosen
D
,
O’Brien
KD
,
O’Rourke
B
,
Park
J-Y
,
Ping
P
,
Sack
MN
,
Sheu
S-S
,
Shi
Y
,
Shiva
S
,
Wallace
DC
,
Weiss
RG
,
Vernon
HJ
,
Wong
R
,
Longacre
LS
,
Shi
S
.
Unlocking the secrets of mitochondria in the cardiovascular system: path to a cure in heart failure—a report from the 2018 national heart, lung, and blood institute workshop
.
Circulation
2019
;
140
:
1205
1216
.

2

Lopaschuk
GD
,
Karwi
QG
,
Tian
R
,
Wende
AR
,
Abel
ED
.
Cardiac energy metabolism in heart failure
.
Circ Res
2021
;
128
:
1487
1513
.

3

Steenbergen
C
,
Murphy
E
,
Watts
JA
,
London
RE
.
Correlation between cytosolic free calcium, contracture, ATP, and irreversible ischemic injury in perfused rat heart
.
Circ Res
1990
;
66
:
135
146
.

4

Bauer
TM
,
Giles
AV
,
Sun
J
,
Femnou
A
,
Covian
R
,
Murphy
E
,
Balaban
RS
.
Perfused murine heart optical transmission spectroscopy using optical catheter and integrating sphere: effects of ischemia/reperfusion
.
Anal Biochem
2019
;
586
:
113443
.

5

Jennings
RB
,
Reimer
KA
,
Steenbergen
C
.
Effect of inhibition of the mitochondrial ATPase on net myocardial ATP in total ischemia
.
J Mol Cell Cardiol
1991
;
23
:
1383
1395
.

6

Chinopoulos
C
.
Mitochondrial consumption of cytosolic ATP: not so fast
.
FEBS Lett
2011
;
585
:
1255
1259
.

7

Ritterhoff
J
,
Tian
R
.
Metabolism in cardiomyopathy: every substrate matters
.
Cardiovasc Res
2017
;
113
:
411
421
.

8

Dennis
SC
,
Gevers
W
,
Opie
LH
.
Protons in ischemia: where do they come from; where do they go to?
J Mol Cell Cardiol
1991
;
23
:
1077
1086
.

9

Murphy
E
,
Perlman
M
,
London
RE
,
Steenbergen
C
.
Amiloride delays the ischemia-induced rise in cytosolic free calcium
.
Circ Res
1991
;
68
:
1250
1258
.

10

Lopaschuk
GD
,
Ussher
JR
,
Folmes
CDL
,
Jaswal
JS
,
Stanley
WC
.
Myocardial fatty acid metabolism in health and disease
.
Physiol Rev
2010
;
90
:
207
258
.

11

Kudo
N
,
Barr
AJ
,
Barr
RL
,
Desai
S
,
Lopaschuk
GD
.
High rates of fatty acid oxidation during reperfusion of ischemic hearts are associated with a decrease in malonyl-CoA levels due to an increase in 5’-AMP-activated protein kinase inhibition of acetyl-CoA carboxylase
.
J Biol Chem
1995
;
270
:
17513
17520
.

12

Evans
RD
,
Clarke
K
.
Myocardial substrate metabolism in heart disease
.
Front Biosci (Schol Ed)
2012
;
4
:
556
580
.

13

Zhou
L
,
Huang
H
,
McElfresh
TA
,
Prosdocimo
DA
,
Stanley
WC
.
Impact of anaerobic glycolysis and oxidative substrate selection on contractile function and mechanical efficiency during moderate severity ischemia
.
Am J Physiol Heart Circ Physiol
2008
;
295
:
H939
H945
.

14

Neubauer
S
,
Horn
M
,
Cramer
M
,
Harre
K
,
Newell
JB
,
Peters
W
,
Pabst
T
,
Ertl
G
,
Hahn
D
,
Ingwall
JS
,
Kochsiek
K
.
Myocardial phosphocreatine-to-ATP ratio is a predictor of mortality in patients with dilated cardiomyopathy
.
Circulation
1997
;
96
:
2190
2196
.

15

Horton
JL
,
Davidson
MT
,
Kurishima
C
,
Vega
RB
,
Powers
JC
,
Matsuura
TR
,
Petucci
C
,
Lewandowski
ED
,
Crawford
PA
,
Muoio
DM
,
Recchia
FA
,
Kelly
DP
.
The failing heart utilizes 3-hydroxybutyrate as a metabolic stress defense
.
JCI Insight
2019
;
4
:
124079
.

16

Kolwicz
SC
.
Ketone body metabolism in the ischemic heart
.
Front Cardiovasc Med
2021
;
8
:
789458
.

17

Ho
KL
,
Zhang
L
,
Wagg
C
,
Al Batran
R
,
Gopal
K
,
Levasseur
J
,
Leone
T
,
Dyck
JRB
,
Ussher
JR
,
Muoio
DM
,
Kelly
DP
,
Lopaschuk
GD
.
Increased ketone body oxidation provides additional energy for the failing heart without improving cardiac efficiency
.
Cardiovasc Res
2019
;
115
:
1606
1616
.

18

Butler
J
,
Hamo
CE
,
Filippatos
G
,
Pocock
SJ
,
Bernstein
RA
,
Brueckmann
M
,
Cheung
AK
,
George
JT
,
Green
JB
,
Januzzi
JL
,
Kaul
S
,
Lam
CSP
,
Lip
GYH
,
Marx
N
,
McCullough
PA
,
Mehta
CR
,
Ponikowski
P
,
Rosenstock
J
,
Sattar
N
,
Salsali
A
,
Scirica
BM
,
Shah
SJ
,
Tsutsui
H
,
Verma
S
,
Wanner
C
,
Woerle
H-J
,
Zannad
F
,
Anker
SD
,
Trials Program EMPEROR
.
The potential role and rationale for treatment of heart failure with sodium-glucose co-transporter 2 inhibitors
.
Eur J Heart Fail
2017
;
19
:
1390
1400
.

19

Sack
MN
,
Yellon
DM
.
Insulin therapy as an adjunct to reperfusion after acute coronary ischemia: a proposed direct myocardial cell survival effect independent of metabolic modulation
.
J Am Coll Cardiol
2003
;
41
:
1404
1407
.

20

Anagnostis
P
,
Athyros
VG
,
Adamidou
F
,
Panagiotou
A
,
Kita
M
,
Karagiannis
A
,
Mikhailidis
DP
.
Glucagon-like peptide-1-based therapies and cardiovascular disease: looking beyond glycaemic control
.
Diabetes Obes Metab
2011
;
13
:
302
312
.

21

Kobara
M
,
Toba
H
,
Nakata
T
.
A glucagon-like peptide 1 analog protects mitochondria and attenuates hypoxia-reoxygenation injury in cultured cardiomyocytes
.
J Cardiovasc Pharmacol
2022
;
79
:
568
576
.

22

Diguet
N
,
Trammell
SAJ
,
Tannous
C
,
Deloux
R
,
Piquereau
J
,
Mougenot
N
,
Gouge
A
,
Gressette
M
,
Manoury
B
,
Blanc
J
,
Breton
M
,
Decaux
J-F
,
Lavery
GG
,
Baczkó
I
,
Zoll
J
,
Garnier
A
,
Li
Z
,
Brenner
C
,
Mericskay
M
.
Nicotinamide riboside preserves cardiac function in a mouse model of dilated cardiomyopathy
.
Circulation
2018
;
137
:
2256
2273
.

23

Walker
MA
,
Tian
R
.
Raising NAD in heart failure: time to translate?
Circulation
2018
;
137
:
2274
2277
.

24

Tong
D
,
Schiattarella
GG
,
Jiang
N
,
Altamirano
F
,
Szweda
PA
,
Elnwasany
A
,
Lee
DI
,
Yoo
H
,
Kass
DA
,
Szweda
LI
,
Lavandero
S
,
Verdin
E
,
Gillette
TG
,
Hill
JA
.
NAD+ repletion reverses heart failure with preserved ejection fraction
.
Circ Res
2021
;
128
:
1629
1641
.

25

Glancy
B
,
Balaban
RS
.
Role of mitochondrial Ca2+ in the regulation of cellular energetics
.
Biochemistry
2012
;
51
:
2959
2973
.

26

Denton
RM
,
McCormack
JG
.
The role of calcium in the regulation of mitochondrial metabolism
.
Biochem Soc Trans
1980
;
8
:
266
268
.

27

Deluca
HF
,
Engstrom
GW
.
Calcium uptake by rat kidney mitochondria
.
Proc Natl Acad Sci U S A
1961
;
47
:
1744
1750
.

28

Brandes
R
,
Bers
DM
.
Simultaneous measurements of mitochondrial NADH and Ca(2+) during increased work in intact rat heart trabeculae
.
Biophys J
2002
;
83
:
587
604
.

29

Liu
T
,
O’Rourke
B
.
Enhancing mitochondrial Ca2+ uptake in myocytes from failing hearts restores energy supply and demand matching
.
Circ Res
2008
;
103
:
279
288
.

30

Wescott
AP
,
Kao
JPY
,
Lederer
WJ
,
Boyman
L
.
Voltage-energized calcium-sensitive ATP production by mitochondria
.
Nat Metab
2019
;
1
:
975
984
.

31

Del Re
DP
,
Amgalan
D
,
Linkermann
A
,
Liu
Q
,
Kitsis
RN
.
Fundamental mechanisms of regulated cell death and implications for heart disease
.
Physiol Rev
2019
;
99
:
1765
1817
.

32

Diao
RY
,
Gustafsson
ÅB
.
Mitochondrial quality surveillance: mitophagy in cardiovascular health and disease
.
Am J Physiol Cell Physiol
2022
;
322
:
C218
C230
.

33

Ikeda
S
,
Zablocki
D
,
Sadoshima
J
.
The role of autophagy in death of cardiomyocytes
.
J Mol Cell Cardiol
2022
;
165
:
1
8
.

34

Martens
MD
,
Karch
J
,
Gordon
JW
.
The molecular mosaic of regulated cell death in the cardiovascular system
.
Biochim Biophys Acta Mol Basis Dis
2022
;
1868
:
166297
.

35

Lemasters
JJ
,
Theruvath
TP
,
Zhong
Z
,
Nieminen
A-L
.
Mitochondrial calcium and the permeability transition in cell death
.
Biochim Biophys Acta
2009
;
1787
:
1395
1401
.

36

Griffiths
EJ
,
Halestrap
AP
.
Mitochondrial non-specific pores remain closed during cardiac ischaemia, but open upon reperfusion
.
Biochem J
1995
;
307
(
Pt 1
):
93
98
.

37

Kristian
T
,
Bernardi
P
,
Siesjö
BK
.
Acidosis promotes the permeability transition in energized mitochondria: implications for reperfusion injury
.
J Neurotrauma
2001
;
18
:
1059
1074
.

38

Bauer
TM
,
Murphy
E
.
Role of mitochondrial calcium and the permeability transition pore in regulating cell death
.
Circ Res
2020
;
126
:
280
293
.

39

Mnatsakanyan
N
,
Jonas
EA
.
ATP synthase c-subunit ring as the channel of mitochondrial permeability transition: regulator of metabolism in development and degeneration
.
J Mol Cell Cardiol
2020
;
144
:
109
118
.

40

Bround
MJ
,
Bers
DM
,
Molkentin
JD
.
A 20/20 view of ANT function in mitochondrial biology and necrotic cell death
.
J Mol Cell Cardiol
2020
;
144
:
A3
A13
.

41

Bernardi
P
,
Carraro
M
,
Lippe
G
.
The mitochondrial permeability transition: recent progress and open questions
.
FEBS J
2022
;
289
:
7051
7074
.

42

Giorgio
V
,
von Stockum
S
,
Antoniel
M
,
Fabbro
A
,
Fogolari
F
,
Forte
M
,
Glick
GD
,
Petronilli
V
,
Zoratti
M
,
Szabó
I
,
Lippe
G
,
Bernardi
P
.
Dimers of mitochondrial ATP synthase form the permeability transition pore
.
Proc Natl Acad Sci U S A
2013
;
110
:
5887
5892
.

43

Alavian
KN
,
Beutner
G
,
Lazrove
E
,
Sacchetti
S
,
Park
H-A
,
Licznerski
P
,
Li
H
,
Nabili
P
,
Hockensmith
K
,
Graham
M
,
Porter
GA
,
Jonas
EA
.
An uncoupling channel within the c-subunit ring of the F1FO ATP synthase is the mitochondrial permeability transition pore
.
Proc Natl Acad Sci U S A
2014
;
111
:
10580
10585
.

44

Carroll
J
,
He
J
,
Ding
S
,
Fearnley
IM
,
Walker
JE
.
Persistence of the permeability transition pore in human mitochondria devoid of an assembled ATP synthase
.
Proc Natl Acad Sci U S A
2019
;
116
:
12816
12821
.

45

He
J
,
Carroll
J
,
Ding
S
,
Fearnley
IM
,
Walker
JE
.
Permeability transition in human mitochondria persists in the absence of peripheral stalk subunits of ATP synthase
.
Proc Natl Acad Sci U S A
2017
;
114
:
9086
9091
.

46

Quôc K
L
,
Quôc D
L
.
Involvement of the ADP/ATP carrier in calcium-induced perturbations of the mitochondrial inner membrane permeability: importance of the orientation of the nucleotide binding site
.
Arch Biochem Biophys
1988
;
265
:
249
257
.

47

Halestrap
A
,
Brenner
C
.
The adenine nucleotide translocase: a central component of the mitochondrial permeability transition pore and key player in cell death
.
CMC
2003
;
10
:
1507
1525
.

48

Kokoszka
JE
,
Waymire
KG
,
Levy
SE
,
Sligh
JE
,
Cai
J
,
Jones
DP
,
MacGregor
GR
,
Wallace
DC
.
The ADP/ATP translocator is not essential for the mitochondrial permeability transition pore
.
Nature
2004
;
427
:
461
465
.

49

Karch
J
,
Bround
MJ
,
Khalil
H
,
Sargent
MA
,
Latchman
N
,
Terada
N
,
Peixoto
PM
,
Molkentin
JD
.
Inhibition of mitochondrial permeability transition by deletion of the ANT family and CypD
.
Sci Adv
2019
;
5
:
eaaw4597
.

50

Giorgio
V
,
Burchell
V
,
Schiavone
M
,
Bassot
C
,
Minervini
G
,
Petronilli
V
,
Argenton
F
,
Forte
M
,
Tosatto
S
,
Lippe
G
,
Bernardi
P
.
Ca2 + binding to F-ATP synthase β subunit triggers the mitochondrial permeability transition
.
EMBO Rep
2017
;
18
:
1065
1076
.

51

Brustovetsky
N
,
Klingenberg
M
.
Mitochondrial ADP/ATP carrier can be reversibly converted into a large channel by Ca2+
.
Biochemistry
1996
;
35
:
8483
8488
.

52

Amanakis
G
,
Sun
J
,
Fergusson
MM
,
McGinty
S
,
Liu
C
,
Molkentin
JD
,
Murphy
E
.
Cysteine 202 of cyclophilin D is a site of multiple post-translational modifications and plays a role in cardioprotection
.
Cardiovasc Res
2021
;
117
:
212
223
.

53

Li
L
,
Thompson
J
,
Hu
Y
,
Lesnefsky
EJ
,
Willard
B
,
Chen
Q
.
Calpain-mediated protein targets in cardiac mitochondria following ischemia-reperfusion
.
Sci Rep
2022
;
12
:
138
.

54

Chen
Q
,
Thompson
J
,
Hu
Y
,
Lesnefsky
EJ
.
Reversing mitochondrial defects in aged hearts: role of mitochondrial calpain activation
.
Am J Physiol Cell Physiol
2022
;
322
:
C296
C310
.

55

Fernandez-Sanz
C
,
Ruiz-Meana
M
,
Castellano
J
,
Miro-Casas
E
,
Nuñez
E
,
Inserte
J
,
Vázquez
J
,
Garcia-Dorado
D
.
Altered FoF1 ATP synthase and susceptibility to mitochondrial permeability transition pore during ischaemia and reperfusion in aging cardiomyocytes
.
Thromb Haemost
2015
;
113
:
441
451
.

56

Andrienko
TN
,
Picht
E
,
Bers
DM
.
Mitochondrial free calcium regulation during sarcoplasmic reticulum calcium release in rat cardiac myocytes
.
J Mol Cell Cardiol
2009
;
46
:
1027
1036
.

57

Boyman
L
,
Chikando
AC
,
Williams
GSB
,
Khairallah
RJ
,
Kettlewell
S
,
Ward
CW
,
Smith
GL
,
Kao
JPY
,
Lederer
WJ
.
Calcium movement in cardiac mitochondria
.
Biophys J
2014
;
107
:
1289
1301
.

58

Fernandez-Sanz
C
,
De la Fuente
S
,
Sheu
S-S
.
Mitochondrial Ca2+ concentrations in live cells: quantification methods and discrepancies
.
FEBS Lett
2019
;
593
:
1528
1541
.

59

O’Rourke
B
,
Blatter
LA
.
Mitochondrial Ca2 + uptake: tortoise or hare?
J Mol Cell Cardiol
2009
;
46
:
767
774
.

60

Williams
GSB
,
Boyman
L
,
Chikando
AC
,
Khairallah
RJ
,
Lederer
WJ
.
Mitochondrial calcium uptake
.
Proc Natl Acad Sci U S A
2013
;
110
:
10479
10486
.

61

Théroux
P
,
Chaitman
BR
,
Danchin
N
,
Erhardt
L
,
Meinertz
T
,
Schroeder
JS
,
Tognoni
G
,
White
HD
,
Willerson
JT
,
Jessel
A
.
Inhibition of the sodium-hydrogen exchanger with cariporide to prevent myocardial infarction in high-risk ischemic situations. Main results of The Guardian trial. Guard during ischemia against necrosis (GUARDIAN) investigators
.
Circulation
2000
;
102
:
3032
3038
.

62

Murphy
E
,
Allen
DG
.
Why did the NHE inhibitor clinical trials fail?
J Mol Cell Cardiol
2009
;
46
:
137
141
.

63

Schäfer
C
,
Ladilov
Y
,
Inserte
J
,
Schäfer
M
,
Haffner
S
,
Garcia-Dorado
D
,
Piper
HM
.
Role of the reverse mode of the Na+/Ca2+ exchanger in reoxygenation-induced cardiomyocyte injury
.
Cardiovasc Res
2001
;
51
:
241
250
.

64

Inserte
J
,
Garcia-Dorado
D
,
Ruiz-Meana
M
,
Padilla
F
,
Barrabés
JA
,
Pina
P
,
Agulló
L
,
Piper
HM
,
Soler-Soler
J
.
Effect of inhibition of Na(+)/Ca(2+) exchanger at the time of myocardial reperfusion on hypercontracture and cell death
.
Cardiovasc Res
2002
;
55
:
739
748
.

65

Jones
SP
,
Tang
X-L
,
Guo
Y
,
Steenbergen
C
,
Lefer
DJ
,
Kukreja
RC
,
Kong
M
,
Li
Q
,
Bhushan
S
,
Zhu
X
,
Du
J
,
Nong
Y
,
Stowers
HL
,
Kondo
K
,
Hunt
GN
,
Goodchild
TT
,
Orr
A
,
Chang
CC
,
Ockaili
R
,
Salloum
FN
,
Bolli
R
.
The NHLBI-sponsored consortium for preclinicAl assESsment of cARdioprotective therapies (CAESAR): a new paradigm for rigorous, accurate, and reproducible evaluation of putative infarct-sparing interventions in mice, rabbits, and pigs
.
Circ Res
2015
;
116
:
572
586
.

66

Lecour
S
,
Andreadou
I
,
Bøtker
HE
,
Davidson
SM
,
Heusch
G
,
Ruiz-Meana
M
,
Schulz
R
,
Zuurbier
CJ
,
Ferdinandy
P
,
Hausenloy
DJ
, on behalf of the
European union-CARDIOPROTECTION COST ACTION CA16225
.
IMproving preclinical assessment of cardioprotective therapies (IMPACT) criteria: guidelines of the EU-CARDIOPROTECTION COST action
.
Basic Res Cardiol
2021
;
116
:
52
.

67

Antoniel
M
,
Jones
K
,
Antonucci
S
,
Spolaore
B
,
Fogolari
F
,
Petronilli
V
,
Giorgio
V
,
Carraro
M
,
Di Lisa
F
,
Forte
M
,
Szabó
I
,
Lippe
G
,
Bernardi
P
.
The unique histidine in OSCP subunit of F-ATP synthase mediates inhibition of the permeability transition pore by acidic pH
.
EMBO Rep
2018
;
19
:
257
268
.

68

Griffiths
EJ
,
Halestrap
AP
.
Protection by cyclosporin A of ischemia/reperfusion-induced damage in isolated rat hearts
.
J Mol Cell Cardiol
1993
;
25
:
1461
1469
.

69

Baines
CP
,
Kaiser
RA
,
Purcell
NH
,
Blair
NS
,
Osinska
H
,
Hambleton
MA
,
Brunskill
EW
,
Sayen
MR
,
Gottlieb
RA
,
Dorn
GW
,
Robbins
J
,
Molkentin
JD
.
Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death
.
Nature
2005
;
434
:
658
662
.

70

Nakagawa
T
,
Shimizu
S
,
Watanabe
T
,
Yamaguchi
O
,
Otsu
K
,
Yamagata
H
,
Inohara
H
,
Kubo
T
,
Tsujimoto
Y
.
Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death
.
Nature
2005
;
434
:
652
658
.

71

Piot
C
,
Croisille
P
,
Staat
P
,
Thibault
H
,
Rioufol
G
,
Mewton
N
,
Elbelghiti
R
,
Cung
TT
,
Bonnefoy
E
,
Angoulvant
D
,
Macia
C
,
Raczka
F
,
Sportouch
C
,
Gahide
G
,
Finet
G
,
André-Fouët
X
,
Revel
D
,
Kirkorian
G
,
Monassier
J-P
,
Derumeaux
G
,
Ovize
M
.
Effect of cyclosporine on reperfusion injury in acute myocardial infarction
.
N Engl J Med
2008
;
359
:
473
481
.

72

Cung
T-T
,
Morel
O
,
Cayla
G
,
Rioufol
G
,
Garcia-Dorado
D
,
Angoulvant
D
,
Bonnefoy-Cudraz
E
,
Guérin
P
,
Elbaz
M
,
Delarche
N
,
Coste
P
,
Vanzetto
G
,
Metge
M
,
Aupetit
J-F
,
Jouve
B
,
Motreff
P
,
Tron
C
,
Labeque
J-N
,
Steg
PG
,
Cottin
Y
,
Range
G
,
Clerc
J
,
Claeys
MJ
,
Coussement
P
,
Prunier
F
,
Moulin
F
,
Roth
O
,
Belle
L
,
Dubois
P
,
Barragan
P
,
Gilard
M
,
Piot
C
,
Colin
P
,
De Poli
F
,
Morice
M-C
,
Ider
O
,
Dubois-Randé
J-L
,
Unterseeh
T
,
Le Breton
H
,
Béard
T
,
Blanchard
D
,
Grollier
G
,
Malquarti
V
,
Staat
P
,
Sudre
A
,
Elmer
E
,
Hansson
MJ
,
Bergerot
C
,
Boussaha
I
,
Jossan
C
,
Derumeaux
G
,
Mewton
N
,
Ovize
M
.
Cyclosporine before PCI in patients with acute myocardial infarction
.
N Engl J Med
2015
;
373
:
1021
1031
.

73

Basso
E
,
Fante
L
,
Fowlkes
J
,
Petronilli
V
,
Forte
MA
,
Bernardi
P
.
Properties of the permeability transition pore in mitochondria devoid of cyclophilin D
.
J Biol Chem
2005
;
280
:
18558
18561
.

74

Bernardi
P
,
Di Lisa
F
.
Cyclosporine before PCI in acute myocardial infarction
.
N Engl J Med
2016
;
374
:
89
90
.

75

Broekemeier
KM
,
Pfeiffer
DR
.
Cyclosporin A-sensitive and insensitive mechanisms produce the permeability transition in mitochondria
.
Biochem Biophys Res Commun
1989
;
163
:
561
566
.

76

Halestrap
AP
,
Richardson
AP
.
The mitochondrial permeability transition: a current perspective on its identity and role in ischaemia/reperfusion injury
.
J Mol Cell Cardiol
2015
;
78
:
129
141
.

77

Baughman
JM
,
Perocchi
F
,
Girgis
HS
,
Plovanich
M
,
Belcher-Timme
CA
,
Sancak
Y
,
Bao
XR
,
Strittmatter
L
,
Goldberger
O
,
Bogorad
RL
,
Koteliansky
V
,
Mootha
VK
.
Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter
.
Nature
2011
;
476
:
341
345
.

78

De Stefani
D
,
Raffaello
A
,
Teardo
E
,
Szabò
I
,
Rizzuto
R
.
A forty-kilodalton protein of the inner membrane is the mitochondrial calcium uniporter
.
Nature
2011
;
476
:
336
340
.

79

Márta
K
,
Hasan
P
,
Rodríguez-Prados
M
,
Paillard
M
,
Hajnóczky
G
.
Pharmacological inhibition of the mitochondrial Ca2+ uniporter: relevance for pathophysiology and human therapy
.
J Mol Cell Cardiol
2021
;
151
:
135
144
.

80

Pan
X
,
Liu
J
,
Nguyen
T
,
Liu
C
,
Sun
J
,
Teng
Y
,
Fergusson
MM
,
Rovira
II
,
Allen
M
,
Springer
DA
,
Aponte
AM
,
Gucek
M
,
Balaban
RS
,
Murphy
E
,
Finkel
T
.
The physiological role of mitochondrial calcium revealed by mice lacking the mitochondrial calcium uniporter
.
Nat Cell Biol
2013
;
15
:
1464
1472
.

81

Kwong
JQ
,
Lu
X
,
Correll
RN
,
Schwanekamp
JA
,
Vagnozzi
RJ
,
Sargent
MA
,
York
AJ
,
Zhang
J
,
Bers
DM
,
Molkentin
JD
.
The mitochondrial calcium uniporter selectively matches metabolic output to acute contractile stress in the heart
.
Cell Rep
2015
;
12
:
15
22
.

82

Luongo
TS
,
Lambert
JP
,
Yuan
A
,
Zhang
X
,
Gross
P
,
Song
J
,
Shanmughapriya
S
,
Gao
E
,
Jain
M
,
Houser
SR
,
Koch
WJ
,
Cheung
JY
,
Madesh
M
,
Elrod
JW
.
The mitochondrial calcium uniporter matches energetic supply with cardiac workload during stress and modulates permeability transition
.
Cell Rep
2015
;
12
:
23
34
.

83

Rasmussen
TP
,
Wu
Y
,
Joiner
MA
,
Koval
OM
,
Wilson
NR
,
Luczak
ED
,
Wang
Q
,
Chen
B
,
Gao
Z
,
Zhu
Z
,
Wagner
BA
,
Soto
J
,
McCormick
ML
,
Kutschke
W
,
Weiss
RM
,
Yu
L
,
Boudreau
RL
,
Abel
ED
,
Zhan
F
,
Spitz
DR
,
Buettner
GR
,
Song
L-S
,
Zingman
LV
,
Anderson
ME
.
Inhibition of MCU forces extramitochondrial adaptations governing physiological and pathological stress responses in heart
.
Proc Natl Acad Sci U S A
2015
;
112
:
9129
9134
.

84

Wu
Y
,
Rasmussen
TP
,
Koval
OM
,
Joiner
M-LA
,
Hall
DD
,
Chen
B
,
Luczak
ED
,
Wang
Q
,
Rokita
AG
,
Wehrens
XHT
,
Song
L-S
,
Anderson
ME
.
The mitochondrial uniporter controls fight or flight heart rate increases
.
Nat Commun
2015
;
6
:
6081
.

85

Parks
RJ
,
Menazza
S
,
Holmström
KM
,
Amanakis
G
,
Fergusson
M
,
Ma
H
,
Aponte
AM
,
Bernardi
P
,
Finkel
T
,
Murphy
E
.
Cyclophilin D-mediated regulation of the permeability transition pore is altered in mice lacking the mitochondrial calcium uniporter
.
Cardiovasc Res
2019
;
115
:
385
394
.

86

Ashok
D
,
Papanicolaou
K
,
Liu
T
,
O’Rourke
B
.
Abstract 554: reverse-mode mitochondrial Na+/Ca2+ mitochondrial exchange, not the mcu, is the primary mode of Ca2+ import into the mitochondria during ischemia/reperfusion in neonatal cardiac myocytes
.
Circ Res
2020
;
127
:A554.

87

Kosmach
A
,
Roman
B
,
Sun
J
,
Femnou
A
,
Zhang
F
,
Liu
C
,
Combs
CA
,
Balaban
RS
,
Murphy
E
.
Monitoring mitochondrial calcium and metabolism in the beating MCU-KO heart
.
Cell Rep
2021
;
37
:
109846
.

88

Rizzuto
R
,
Bernardi
P
,
Favaron
M
,
Azzone
GF
.
Pathways for Ca2+ efflux in heart and liver mitochondria
.
Biochem J
1987
;
246
:
271
277
.

89

Palty
R
,
Silverman
WF
,
Hershfinkel
M
,
Caporale
T
,
Sensi
SL
,
Parnis
J
,
Nolte
C
,
Fishman
D
,
Shoshan-Barmatz
V
,
Herrmann
S
,
Khananshvili
D
,
Sekler
I
.
NCLX Is an essential component of mitochondrial Na+/Ca2+ exchange
.
Proc Natl Acad Sci U S A
2010
;
107
:
436
441
.

90

Luongo
TS
,
Lambert
JP
,
Gross
P
,
Nwokedi
M
,
Lombardi
AA
,
Shanmughapriya
S
,
Carpenter
AC
,
Kolmetzky
D
,
Gao
E
,
van Berlo
JH
,
Tsai
EJ
,
Molkentin
JD
,
Chen
X
,
Madesh
M
,
Houser
SR
,
Elrod
JW
.
The mitochondrial Na+/Ca2+ exchanger is essential for Ca2+ homeostasis and viability
.
Nature
2017
;
545
:
93
97
.

91

Piper
HM
,
García-Dorado
D
,
Ovize
M
.
A fresh look at reperfusion injury
.
Cardiovasc Res
1998
;
38
:
291
300
.

92

Garcia-Dorado
D
,
Ruiz-Meana
M
,
Inserte
J
,
Rodriguez-Sinovas
A
,
Piper
HM
.
Calcium-mediated cell death during myocardial reperfusion
.
Cardiovasc Res
2012
;
94
:
168
180
.

93

Santulli
G
,
Xie
W
,
Reiken
SR
,
Marks
AR
.
Mitochondrial calcium overload is a key determinant in heart failure
.
Proc Natl Acad Sci U S A
2015
;
112
:
11389
11394
.

94

Ruiz-Meana
M
,
Minguet
M
,
Bou-Teen
D
,
Miro-Casas
E
,
Castans
C
,
Castellano
J
,
Bonzon-Kulichenko
E
,
Igual
A
,
Rodriguez-Lecoq
R
,
Vázquez
J
,
Garcia-Dorado
D
.
Ryanodine receptor glycation favors mitochondrial damage in the senescent heart
.
Circulation
2019
;
139
:
949
964
.

95

Holmström
KM
,
Pan
X
,
Liu
JC
,
Menazza
S
,
Liu
J
,
Nguyen
TT
,
Pan
H
,
Parks
RJ
,
Anderson
S
,
Noguchi
A
,
Springer
D
,
Murphy
E
,
Finkel
T
.
Assessment of cardiac function in mice lacking the mitochondrial calcium uniporter
.
J Mol Cell Cardiol
2015
;
85
:
178
182
.

96

Puente
BN
,
Sun
J
,
Parks
RJ
,
Fergusson
MM
,
Liu
C
,
Springer
DA
,
Aponte
AM
,
Liu
JC
,
Murphy
E
.
MICU3 plays an important role in cardiovascular function
.
Circ Res
2020
;
127
:
1571
1573
.

97

Kohlhaas
M
,
Maack
C
.
Adverse bioenergetic consequences of Na+–Ca2+ exchanger–mediated Ca2+ influx in cardiac myocytes
.
Circulation
2010
;
122
:
2273
2280
.

98

Liu
T
,
Takimoto
E
,
Dimaano
VL
,
DeMazumder
D
,
Kettlewell
S
,
Smith
G
,
Sidor
A
,
Abraham
TP
,
O’Rourke
B
.
Inhibiting mitochondrial Na+/Ca2+ exchange prevents sudden death in a Guinea pig model of heart failure
.
Circ Res
2014
;
115
:
44
54
.

99

Maack
C
,
Cortassa
S
,
Aon
MA
,
Ganesan
AN
,
Liu
T
,
O’Rourke
B
.
Elevated cytosolic Na+ decreases mitochondrial Ca2+ uptake during excitation–contraction coupling and impairs energetic adaptation in cardiac myocytes
.
Circ Res
2006
;
99
:
172
182
.

100

Kohlhaas
M
,
Liu
T
,
Knopp
A
,
Zeller
T
,
Ong
MF
,
Böhm
M
,
O’Rourke
B
,
Maack
C
.
Elevated cytosolic Na+ increases mitochondrial formation of reactive oxygen species in failing cardiac myocytes
.
Circulation
2010
;
121
:
1606
1613
.

101

Liu
T
,
Yang
N
,
Sidor
A
,
O’Rourke
B
.
MCU overexpression rescues inotropy and reverses heart failure by reducing SR Ca2+ leak
.
Circ Res
2021
;
128
:
1191
1204
.

102

Boengler
K
,
Stahlhofen
S
,
van de Sand
A
,
Gres
P
,
Ruiz-Meana
M
,
Garcia-Dorado
D
,
Heusch
G
,
Schulz
R
.
Presence of connexin 43 in subsarcolemmal, but not in interfibrillar cardiomyocyte mitochondria
.
Basic Res Cardiol
2009
;
104
:
141
147
.

103

Ruiz-Meana
M
,
Rodríguez-Sinovas
A
,
Cabestrero
A
,
Boengler
K
,
Heusch
G
,
Garcia-Dorado
D
.
Mitochondrial connexin43 as a new player in the pathophysiology of myocardial ischaemia-reperfusion injury
.
Cardiovasc Res
2008
;
77
:
325
333
.

104

Garbincius
JF
,
Elrod
JW
.
Is the failing heart starved of mitochondrial calcium?
Circ Res
2021
;
128
:
1205
1207
.

105

Brand
MD
.
Mitochondrial generation of superoxide and hydrogen peroxide as the source of mitochondrial redox signaling
.
Free Radic Biol Med
2016
;
100
:
14
31
.

106

Orr
AL
,
Quinlan
CL
,
Perevoshchikova
IV
,
Brand
MD
.
A refined analysis of superoxide production by mitochondrial sn-glycerol 3-phosphate dehydrogenase
.
J Biol Chem
2012
;
287
:
42921
42935
.

107

Crochemore
C
,
Mekki
M
,
Corbière
C
,
Karoui
A
,
Noël
R
,
Vendeville
C
,
Vaugeois
J-M
,
Monteil
C
.
Subsarcolemmal and interfibrillar mitochondria display distinct superoxide production profiles
.
Free Radic Res
2015
;
49
:
331
337
.

108

Murphy
MP
.
How mitochondria produce reactive oxygen species
.
Biochem J
2009
;
417
:
1
13
.

109

Balaban
RS
,
Nemoto
S
,
Finkel
T
.
Mitochondria, oxidants, and aging
.
Cell
2005
;
120
:
483
495
.

110

Wong
H-S
,
Dighe
PA
,
Mezera
V
,
Monternier
P-A
,
Brand
MD
.
Production of superoxide and hydrogen peroxide from specific mitochondrial sites under different bioenergetic conditions
.
J Biol Chem
2017
;
292
:
16804
16809
.

111

Peoples
JN
,
Saraf
A
,
Ghazal
N
,
Pham
TT
,
Kwong
JQ
.
Mitochondrial dysfunction and oxidative stress in heart disease
.
Exp Mol Med
2019
;
51
:
1
13
.

112

Hamanaka
RB
,
Chandel
NS
.
Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes
.
Trends Biochem Sci
2010
;
35
:
505
513
.

113

Sugamura
K
,
Keaney
JF
.
Reactive oxygen species in cardiovascular disease
.
Free Radic Biol Med
2011
;
51
:
978
992
.

114

Zorov
DB
,
Filburn
CR
,
Klotz
LO
,
Zweier
JL
,
Sollott
SJ
.
Reactive oxygen species (ROS)-induced ROS release: a new phenomenon accompanying induction of the mitochondrial permeability transition in cardiac myocytes
.
J Exp Med
2000
;
192
:
1001
1014
.

115

Zorov
DB
,
Juhaszova
M
,
Sollott
SJ
.
Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release
.
Physiol Rev
2014
;
94
:
909
950
.

116

Brand
MD
.
Riding the tiger – physiological and pathological effects of superoxide and hydrogen peroxide generated in the mitochondrial matrix
.
Crit Rev Biochem Mol Biol
2020
;
55
:
592
661
.

117

Castro
L
,
Tórtora
V
,
Mansilla
S
,
Radi
R
.
Aconitases: non-redox iron-Sulfur proteins sensitive to reactive Species
.
Acc Chem Res
2019
;
52
:
2609
2619
.

118

Murphy
MP
.
Mitochondrial thiols in antioxidant protection and redox signaling: distinct roles for glutathionylation and other thiol modifications
.
Antioxid Redox Signal
2012
;
16
:
476
495
.

119

Radi
R
,
Turrens
JF
,
Chang
LY
,
Bush
KM
,
Crapo
JD
,
Freeman
BA
.
Detection of catalase in rat heart mitochondria
.
J Biol Chem
1991
;
266
:
22028
22034
.

120

Rindler
PM
,
Plafker
SM
,
Szweda
LI
,
Kinter
M
.
High dietary fat selectively increases catalase expression within cardiac mitochondria
.
J Biol Chem
2013
;
288
:
1979
1990
.

121

Zhang
X
,
Ji
R
,
Liao
X
,
Castillero
E
,
Kennel
PJ
,
Brunjes
DL
,
Franz
M
,
Möbius-Winkler
S
,
Drosatos
K
,
George
I
,
Chen
EI
,
Colombo
PC
,
Schulze
PC
.
MicroRNA-195 regulates metabolism in failing myocardium via alterations in sirtuin 3 expression and mitochondrial protein acetylation
.
Circulation
2018
;
137
:
2052
2067
.

122

Asimakis
GK
,
Lick
S
,
Patterson
C
.
Postischemic recovery of contractile function is impaired in SOD2(+/−) but not SOD1(+/−) mouse hearts
.
Circulation
2002
;
105
:
981
986
.

123

Chen
Z
,
Siu
B
,
Ho
YS
,
Vincent
R
,
Chua
CC
,
Hamdy
RC
,
Chua
BH
.
Overexpression of MnSOD protects against myocardial ischemia/reperfusion injury in transgenic mice
.
J Mol Cell Cardiol
1998
;
30
:
2281
2289
.

124

Loor
G
,
Kondapalli
J
,
Iwase
H
,
Chandel
NS
,
Waypa
GB
,
Guzy
RD
,
Vanden Hoek
TL
,
Schumacker
PT
.
Mitochondrial oxidant stress triggers cell death in simulated ischemia-reperfusion
.
Biochim Biophys Acta
2011
;
1813
:
1382
1394
.

125

Brand
MD
,
Goncalves
RLS
,
Orr
AL
,
Vargas
L
,
Gerencser
AA
,
Borch Jensen
M
,
Wang
YT
,
Melov
S
,
Turk
CN
,
Matzen
JT
,
Dardov
VJ
,
Petrassi
HM
,
Meeusen
SL
,
Perevoshchikova
IV
,
Jasper
H
,
Brookes
PS
,
Ainscow
EK
.
Suppressors of superoxide-H2O2 production at site IQ of mitochondrial Complex I protect against stem cell hyperplasia and ischemia-reperfusion injury
.
Cell Metab
2016
;
24
:
582
592
.

126

Detaille
D
,
Pasdois
P
,
Sémont
A
,
Dos Santos
P
,
Diolez
P
.
An old medicine as a new drug to prevent mitochondrial complex I from producing oxygen radicals
.
PLoS One
2019
;
14
:
e0216385
.

127

Dai
W
,
Amoedo
ND
,
Perry
J
,
Le Grand
B
,
Boucard
A
,
Carreno
J
,
Zhao
L
,
Brown
DA
,
Rossignol
R
,
Kloner
RA
.
Effects of OP2113 on myocardial infarct size and No reflow in a rat myocardial ischemia/reperfusion model
.
Cardiovasc Drugs Ther
2022
;
36
:
217
227
.

128

Chouchani
ET
,
Pell
VR
,
Gaude
E
,
Aksentijević
D
,
Sundier
SY
,
Robb
EL
,
Logan
A
,
Nadtochiy
SM
,
Ord
ENJ
,
Smith
AC
,
Eyassu
F
,
Shirley
R
,
Hu
C-H
,
Dare
AJ
,
James
AM
,
Rogatti
S
,
Hartley
RC
,
Eaton
S
,
Costa
ASH
,
Brookes
PS
,
Davidson
SM
,
Duchen
MR
,
Saeb-Parsy
K
,
Shattock
MJ
,
Robinson
AJ
,
Work
LM
,
Frezza
C
,
Krieg
T
,
Murphy
MP
.
Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS
.
Nature
2014
;
515
:
431
435
.

129

Valls-Lacalle
L
,
Barba
I
,
Miró-Casas
E
,
Ruiz-Meana
M
,
Rodríguez-Sinovas
A
,
García-Dorado
D
.
Selective inhibition of succinate dehydrogenase in reperfused myocardium with intracoronary malonate reduces infarct size
.
Sci Rep
2018
;
8
:
2442
.

130

Andrienko
TN
,
Pasdois
P
,
Pereira
GC
,
Ovens
MJ
,
Halestrap
AP
.
The role of succinate and ROS in reperfusion injury – a critical appraisal
.
J Mol Cell Cardiol
2017
;
110
:
1
14
.

131

Zhao
K
,
Zhao
G-M
,
Wu
D
,
Soong
Y
,
Birk
AV
,
Schiller
PW
,
Szeto
HH
.
Cell-permeable peptide antioxidants targeted to inner mitochondrial membrane inhibit mitochondrial swelling, oxidative cell death, and reperfusion injury
.
J Biol Chem
2004
;
279
:
34682
34690
.

132

Cho
J
,
Won
K
,
Wu
D
,
Soong
Y
,
Liu
S
,
Szeto
HH
,
Hong
MK
.
Potent mitochondria-targeted peptides reduce myocardial infarction in rats
.
Coron Artery Dis
2007
;
18
:
215
220
.

133

Kloner
RA
,
Hale
SL
,
Dai
W
,
Gorman
RC
,
Shuto
T
,
Koomalsingh
KJ
,
Gorman
JH
,
Sloan
RC
,
Frasier
CR
,
Watson
CA
,
Bostian
PA
,
Kypson
AP
,
Brown
DA
.
Reduction of ischemia/reperfusion injury with bendavia, a mitochondria-targeting cytoprotective peptide
.
J Am Heart Assoc
2012
;
1
:
e001644
.

134

Brown
DA
,
Hale
SL
,
Baines
CP
,
del Rio
CL
,
Hamlin
RL
,
Yueyama
Y
,
Kijtawornrat
A
,
Yeh
ST
,
Frasier
CR
,
Stewart
LM
,
Moukdar
F
,
Shaikh
SR
,
Fisher-Wellman
KH
,
Neufer
PD
,
Kloner
RA
.
Reduction of early reperfusion injury with the mitochondria-targeting peptide bendavia
.
J Cardiovasc Pharmacol Ther
2014
;
19
:
121
132
.

135

Gibson
CM
,
Giugliano
RP
,
Kloner
RA
,
Bode
C
,
Tendera
M
,
Jánosi
A
,
Merkely
B
,
Godlewski
J
,
Halaby
R
,
Korjian
S
,
Daaboul
Y
,
Chakrabarti
AK
,
Spielman
K
,
Neal
BJ
,
Weaver
WD
.
EMBRACE STEMI study: a phase 2a trial to evaluate the safety, tolerability, and efficacy of intravenous MTP-131 on reperfusion injury in patients undergoing primary percutaneous coronary intervention
.
Eur Heart J
2016
;
37
:
1296
1303
.

136

Li
Y
,
Huang
T-T
,
Carlson
EJ
,
Melov
S
,
Ursell
PC
,
Olson
JL
,
Noble
LJ
,
Yoshimura
MP
,
Berger
C
,
Chan
PH
,
Wallace
DC
,
Epstein
CJ
.
Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase
.
Nat Genet
1995
;
11
:
376
381
.

137

Huang
TT
,
Carlson
EJ
,
Gillespie
AM
,
Epstein
CJ
.
Genetic modification of the dilated cardiomyopathy and neonatal lethality phenotype of mice lacking manganese superoxide dismutase
.
Age (Omaha)
1998
;
21
:
83
84
.

138

Lebovitz
RM
,
Zhang
H
,
Vogel
H
,
Cartwright
J
,
Dionne
L
,
Lu
N
,
Huang
S
,
Matzuk
MM
.
Neurodegeneration, myocardial injury, and perinatal death in mitochondrial superoxide dismutase-deficient mice
.
Proc Natl Acad Sci U S A
1996
;
93
:
9782
9787
.

139

Nojiri
H
,
Shimizu
T
,
Funakoshi
M
,
Yamaguchi
O
,
Zhou
H
,
Kawakami
S
,
Ohta
Y
,
Sami
M
,
Tachibana
T
,
Ishikawa
H
,
Kurosawa
H
,
Kahn
RC
,
Otsu
K
,
Shirasawa
T
.
Oxidative stress causes heart failure with impaired mitochondrial respiration
.
J Biol Chem
2006
;
281
:
33789
33801
.

140

Van Remmen
H
,
Williams
MD
,
Guo
Z
,
Estlack
L
,
Yang
H
,
Carlson
EJ
,
Epstein
CJ
,
Huang
TT
,
Richardson
A
.
Knockout mice heterozygous for Sod2 show alterations in cardiac mitochondrial function and apoptosis
.
Am J Physiol Heart Circ Physiol
2001
;
281
:
H1422
H1432
.

141

Strassburger
M
,
Bloch
W
,
Sulyok
S
,
Schüller
J
,
Keist
AF
,
Schmidt
A
,
Wenk
J
,
Peters
T
,
Wlaschek
M
,
Lenart
J
,
Krieg
T
,
Hafner
M
,
Kümin
A
,
Werner
S
,
Müller
W
,
Scharffetter-Kochanek
K
.
Heterozygous deficiency of manganese superoxide dismutase results in severe lipid peroxidation and spontaneous apoptosis in murine myocardium in vivo
.
Free Radic Biol Med
2005
;
38
:
1458
1470
.

142

Almomani
R
,
Herkert
JC
,
Posafalvi
A
,
Post
JG
,
Boven
LG
,
van der Zwaag
PA
,
Willems
PHGM
,
van Veen-Hof
IH
,
Verhagen
JMA
,
Wessels
MW
,
Nikkels
PGJ
,
Wintjes
LT
,
van den Berg
MP
,
Sinke
RJ
,
Rodenburg
RJ
,
Niezen-Koning
KE
,
van Tintelen
JP
,
Jongbloed
JDH
.
Homozygous damaging SOD2 variant causes lethal neonatal dilated cardiomyopathy
.
J Med Genet
2020
;
57
:
23
30
.

143

Kang
PT
,
Chen
C-L
,
Ohanyan
V
,
Luther
DJ
,
Meszaros
JG
,
Chilian
WM
,
Chen
Y-R
.
Overexpressing superoxide dismutase 2 induces a supernormal cardiac function by enhancing redox-dependent mitochondrial function and metabolic dilation
.
J Mol Cell Cardiol
2015
;
88
:
14
28
.

144

Dey
S
,
DeMazumder
D
,
Sidor
A
,
Foster
DB
,
O’Rourke
B
.
Mitochondrial ROS drive sudden cardiac death and chronic proteome remodeling in heart failure
.
Circ Res
2018
;
123
:
356
371
.

145

Hoshino
A
,
Okawa
Y
,
Ariyoshi
M
,
Kaimoto
S
,
Uchihashi
M
,
Fukai
K
,
Iwai-Kanai
E
,
Matoba
S
.
Oxidative post-translational modifications develop LONP1 dysfunction in pressure overload heart failure
.
Circ Heart Fail
2014
;
7
:
500
509
.

146

Adlam
VJ
,
Harrison
JC
,
Porteous
CM
,
James
AM
,
Smith
RAJ
,
Murphy
MP
,
Sammut
IA
.
Targeting an antioxidant to mitochondria decreases cardiac ischemia-reperfusion injury
.
FASEB J
2005
;
19
:
1088
1095
.

147

Neuzil
J
,
Widén
C
,
Gellert
N
,
Swettenham
E
,
Zobalova
R
,
Dong
L-F
,
Wang
X-F
,
Lidebjer
C
,
Dalen
H
,
Headrick
JP
,
Witting
PK
.
Mitochondria transmit apoptosis signalling in cardiomyocyte-like cells and isolated hearts exposed to experimental ischemia-reperfusion injury
.
Redox Rep
2007
;
12
:
148
162
.

148

Ribeiro Junior
RF
,
Dabkowski
ER
,
Shekar
KC
,
O Connell
KA
,
Hecker
PA
,
Murphy
MP
.
Mitoq improves mitochondrial dysfunction in heart failure induced by pressure overload
.
Free Radic Biol Med
2018
;
117
:
18
29
.

149

Goh
KY
,
He
L
,
Song
J
,
Jinno
M
,
Rogers
AJ
,
Sethu
P
,
Halade
GV
,
Rajasekaran
NS
,
Liu
X
,
Prabhu
SD
,
Darley-Usmar
V
,
Wende
AR
,
Zhou
L
.
Mitoquinone ameliorates pressure overload-induced cardiac fibrosis and left ventricular dysfunction in mice
.
Redox Biol
2019
;
21
:
101100
.

150

Kim
S
,
Song
J
,
Ernst
P
,
Latimer
MN
,
Ha
C-M
,
Goh
KY
,
Ma
W
,
Rajasekaran
N-S
,
Zhang
J
,
Liu
X
,
Prabhu
SD
,
Qin
G
,
Wende
AR
,
Young
ME
,
Zhou
L
.
Mitoq regulates redox-related noncoding RNAs to preserve mitochondrial network integrity in pressure-overload heart failure
.
Am J Physiol Heart Circ Physiol
2020
;
318
:
H682
H695
.

151

Snow
BJ
,
Rolfe
FL
,
Lockhart
MM
,
Frampton
CM
,
O’Sullivan
JD
,
Fung
V
,
Smith
RAJ
,
Murphy
MP
,
Taylor
KM
,
Group
PS
.
A double-blind, placebo-controlled study to assess the mitochondria-targeted antioxidant MitoQ as a disease-modifying therapy in Parkinson’s disease
.
Mov Disord
2010
;
25
:
1670
1674
.

152

Gane
EJ
,
Weilert
F
,
Orr
DW
,
Keogh
GF
,
Gibson
M
,
Lockhart
MM
,
Frampton
CM
,
Taylor
KM
,
Smith
RAJ
,
Murphy
MP
.
The mitochondria-targeted anti-oxidant mitoquinone decreases liver damage in a phase II study of hepatitis C patients
.
Liver Int
2010
;
30
:
1019
1026
.

153

Matsushima
S
,
Ide
T
,
Yamato
M
,
Matsusaka
H
,
Hattori
F
,
Ikeuchi
M
,
Kubota
T
,
Sunagawa
K
,
Hasegawa
Y
,
Kurihara
T
,
Oikawa
S
,
Kinugawa
S
,
Tsutsui
H
.
Overexpression of mitochondrial peroxiredoxin-3 prevents left ventricular remodeling and failure after myocardial infarction in mice
.
Circulation
2006
;
113
:
1779
1786
.

154

Dai
D-F
,
Hsieh
EJ
,
Liu
Y
,
Chen
T
,
Beyer
RP
,
Chin
MT
,
MacCoss
MJ
,
Rabinovitch
PS
.
Mitochondrial proteome remodelling in pressure overload-induced heart failure: the role of mitochondrial oxidative stress
.
Cardiovasc Res
2012
;
93
:
79
88
.

155

Sverdlov
AL
,
Elezaby
A
,
Qin
F
,
Behring
JB
,
Luptak
I
,
Calamaras
TD
,
Siwik
DA
,
Miller
EJ
,
Liesa
M
,
Shirihai
OS
,
Pimentel
DR
,
Cohen
RA
,
Bachschmid
MM
,
Colucci
WS
.
Mitochondrial reactive oxygen species mediate cardiac structural, functional, and mitochondrial consequences of diet-induced metabolic heart disease
.
JAHA
2016
;
5
:e002555.

156

Treuting
PM
,
Linford
NJ
,
Knoblaugh
SE
,
Emond
MJ
,
Morton
JF
,
Martin
GM
,
Rabinovitch
PS
,
Ladiges
WC
.
Reduction of age-associated pathology in old mice by overexpression of catalase in mitochondria
.
J Gerontol Ser A: Biol Sci Med Sci
2008
;
63
:
813
822
.

157

Dai
D-F
,
Chen
T
,
Wanagat
J
,
Laflamme
M
,
Marcinek
DJ
,
Emond
MJ
,
Ngo
CP
,
Prolla
TA
,
Rabinovitch
PS
.
Age-dependent cardiomyopathy in mitochondrial mutator mice is attenuated by overexpression of catalase targeted to mitochondria
.
Aging Cell
2010
;
9
:
536
544
.

158

Owada
T
,
Yamauchi
H
,
Saitoh
S-I
,
Miura
S
,
Machii
H
,
Takeishi
Y
.
Resolution of mitochondrial oxidant stress improves aged-cardiovascular performance
.
Coron Artery Dis
2017
;
28
:
33
43
.

159

Olgar
Y
,
Degirmenci
S
,
Durak
A
,
Billur
D
,
Can
B
,
Kayki-Mutlu
G
,
Arioglu-Inan E
E
,
Turan
B
.
Aging related functional and structural changes in the heart and aorta: MitoTEMPO improves aged-cardiovascular performance
.
Exp Gerontol
2018
;
110
:
172
181
.

160

Ni
R
,
Cao
T
,
Xiong
S
,
Ma
J
,
Fan
G-C
,
Lacefield
JC
,
Lu
Y
,
Le Tissier
S
,
Peng
T
.
Therapeutic inhibition of mitochondrial reactive oxygen species with mito-TEMPO reduces diabetic cardiomyopathy
.
Free Radic Biol Med
2016
;
90
:
12
23
.

161

Dai
D-F
,
Hsieh
EJ
,
Chen
T
,
Menendez
LG
,
Basisty
NB
,
Tsai
L
,
Beyer
RP
,
Crispin
DA
,
Shulman
NJ
,
Szeto
HH
,
Tian
R
,
MacCoss
MJ
,
Rabinovitch
PS
.
Global proteomics and pathway analysis of pressure-overload-induced heart failure and its attenuation by mitochondrial-targeted peptides
.
Circ Heart Fail
2013
;
6
:
1067
1076
.

162

Dai
D-F
,
Chen
T
,
Szeto
H
,
Nieves-Cintrón
M
,
Kutyavin
V
,
Santana
LF
,
Rabinovitch
PS
.
Mitochondrial targeted antioxidant peptide ameliorates hypertensive cardiomyopathy
.
J Am Coll Cardiol
2011
;
58
:
73
82
.

163

Sabbah
HN
,
Gupta
RC
,
Kohli
S
,
Wang
M
,
Hachem
S
,
Zhang
K
.
Chronic therapy with elamipretide (MTP-131), a novel mitochondria-targeting peptide, improves left ventricular and mitochondrial function in dogs with advanced heart failure
.
Circ Heart Fail
2016
;
9
:
e002206
.

164

Daubert
MA
,
Yow
E
,
Dunn
G
,
Marchev
S
,
Barnhart
H
,
Douglas
PS
,
O’Connor
C
,
Goldstein
S
,
Udelson
JE
,
Sabbah
HN
.
Novel mitochondria-targeting peptide in heart failure treatment: A randomized, placebo-controlled trial of elamipretide
.
Circ Heart Fail
2017
;
10
:
e004389
.

165

Bertero
E
,
Maack
C
.
Calcium signaling and reactive oxygen Species in mitochondria
.
Circ Res
2018
;
122
:
1460
1478
.

166

Nickel
AG
,
von Hardenberg
A
,
Hohl
M
,
Löffler
JR
,
Kohlhaas
M
,
Becker
J
,
Reil
J-C
,
Kazakov
A
,
Bonnekoh
J
,
Stadelmaier
M
,
Puhl
S-L
,
Wagner
M
,
Bogeski
I
,
Cortassa
S
,
Kappl
R
,
Pasieka
B
,
Lafontaine
M
,
Lancaster
CRD
,
Blacker
TS
,
Hall
AR
,
Duchen
MR
,
Kästner
L
,
Lipp
P
,
Zeller
T
,
Müller
C
,
Knopp
A
,
Laufs
U
,
Böhm
M
,
Hoth
M
,
Maack
C
.
Reversal of mitochondrial transhydrogenase causes oxidative stress in heart failure
.
Cell Metab
2015
;
22
:
472
484
.

167

Zhang
H
,
Limphong
P
,
Pieper
J
,
Liu
Q
,
Rodesch
CK
,
Christians
E
,
Benjamin
IJ
.
Glutathione-dependent reductive stress triggers mitochondrial oxidation and cytotoxicity
.
FASEB J
2012
;
26
:
1442
1451
.

168

Rajasekaran
NS
,
Connell
P
,
Christians
ES
,
Yan
L-J
,
Taylor
RP
,
Orosz
A
,
Zhang
XQ
,
Stevenson
TJ
,
Peshock
RM
,
Leopold
JA
,
Barry
WH
,
Loscalzo
J
,
Odelberg
SJ
,
Benjamin
IJ
.
Human alpha B-crystallin mutation causes oxido-reductive stress and protein aggregation cardiomyopathy in mice
.
Cell
2007
;
130
:
427
439
.

169

Atashi
F
,
Modarressi
A
,
Pepper
MS
.
The role of reactive oxygen species in mesenchymal stem cell adipogenic and osteogenic differentiation: a review
.
Stem Cells Dev
2015
;
24
:
1150
1163
.

170

Song
M
,
Chen
Y
,
Gong
G
,
Murphy
E
,
Rabinovitch
PS
,
Dorn
GW
.
Super-suppression of mitochondrial reactive oxygen species signaling impairs compensatory autophagy in primary mitophagic cardiomyopathy
.
Circ Res
2014
;
115
:
348
353
.

171

Yun
J
,
Finkel
T
.
Mitohormesis
.
Cell Metab
2014
;
19
:
757
766
.

172

Lonn
E
,
Bosch
J
,
Yusuf
S
,
Sheridan
P
,
Pogue
J
,
Arnold
JMO
,
Ross
C
,
Arnold
A
,
Sleight
P
,
Probstfield
J
,
Dagenais
GR
,
HOPE and HOPE-TOO Trial Investigators
.
Effects of long-term vitamin E supplementation on cardiovascular events and cancer: a randomized controlled trial
.
JAMA
2005
;
293
:
1338
1347
.

Author notes

Conflict of interest: None declared.

This work is written by (a) US Government employee(s) and is in the public domain in the US.